Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 24(1): 124, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38267866

RESUMEN

HLX01 (HanliKang®) is a rituximab biosimilar that showed bioequivalence to reference rituximab in untreated CD20-positive diffuse large B-cell lymphoma (DLBCL) in the phase 3 HLX01-NHL03 study. Here, we report the 5-year follow-up results from the open-label extension part. Patients were randomised to either rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) or HLX01 plus CHOP (H-CHOP) every 21 days for up to six cycles. The primary efficacy endpoint was overall survival (OS), and secondary efficacy endpoint was progression-free survival (PFS). Of the 407 patients enrolled in HLX01-NHL03, 316 patients (H-CHOP = 157; R-CHOP = 159) were included in the 5-year follow-up for a median duration of 65.1 (range, 2.2-76.5) months. 96.5% of the patients had an International Prognostic Index (IPI) of 1 or 2, and 17.7% had bone marrow involvement. The 5-year OS rates were 81.0% (95% CI: 74.9-87.5%) and 75.4% (95% CI: 68.9-82.6%)( HR: 0.75, 95% CI 0.47-1.20; p = 0.23) while 5-year PFS rates were 77.7% (95% CI: 71.4-84.6%) and 73.0% (95% CI: 66.3-80.3%) (HR: 0.84, 95% CI 0.54-1.30; p = 0.43) in the H-CHOP and R-CHOP groups, respectively. Treatment outcomes did not differ between groups regardless of IPI score and were consistent with the primary analysis. H-CHOP and R-CHOP provided no significant difference in 5-year OS or PFS in previously untreated patients with low or low-intermediate risk DLBCL.


Asunto(s)
Biosimilares Farmacéuticos , Linfoma de Células B Grandes Difuso , Humanos , Biosimilares Farmacéuticos/efectos adversos , Rituximab/efectos adversos , Estudios de Seguimiento , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Ciclofosfamida/efectos adversos , Doxorrubicina , Prednisona/efectos adversos
2.
Cell Commun Signal ; 22(1): 303, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831321

RESUMEN

BACKGROUND: While previous studies have primarily focused on Glucose transporter type 1 (GLUT1) related glucose metabolism signaling, we aim to discover if GLUT1 promotes tumor progression through a non-metabolic pathway. METHODS: The RNA-seq and microarray data were comprehensively analyzed to evaluate the significance of GLUT1 expression in lung adenocarcinoma (LUAD). The cell proliferation, colony formation, invasion, and migration were used to test GLUT1 's oncogenic function. Co-immunoprecipitation and mass spectrum (MS) were used to uncover potential GLUT1 interacting proteins. RNA-seq, DIA-MS, western blot, and qRT-PCR to probe the change of gene and cell signaling pathways. RESULTS: We found that GLUT1 is highly expressed in LUAD, and higher expression is related to poor patient survival. GLUT1 knockdown caused a decrease in cell proliferation, colony formation, migration, invasion, and induced apoptosis in LUAD cells. Mechanistically, GLUT1 directly interacted with phosphor-epidermal growth factor receptor (p-EGFR) and prevented EGFR protein degradation via ubiquitin-mediated proteolysis. The GLUT1 inhibitor WZB117 can increase the sensitivity of LUAD cells to EGFR-tyrosine kinase inhibitors (TKIs) Gefitinib. CONCLUSIONS: GLUT1 expression is higher in LUAD and plays an oncogenic role in lung cancer progression. Combining GLUT1 inhibitors and EGFR-TKIs could be a potential therapeutic option for LUAD treatment.


Asunto(s)
Adenocarcinoma del Pulmón , Proliferación Celular , Receptores ErbB , Transportador de Glucosa de Tipo 1 , Neoplasias Pulmonares , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética , Humanos , Receptores ErbB/metabolismo , Receptores ErbB/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Fosforilación , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Unión Proteica , Apoptosis , Estabilidad Proteica
3.
Cell Commun Signal ; 21(1): 327, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974196

RESUMEN

Regulated cell death (RCD) is a regulable cell death that involves well-organized signaling cascades and molecular mechanisms. RCD is implicated in fundamental processes such as organ production and tissue remodeling, removing superfluous structures or cells, and regulating cell numbers. Previous studies have not been able to reveal the complete mechanisms, and novel methods of RCD are constantly being proposed. Two metal ions, iron (Fe) and copper (Cu) are essential factors leading to RCDs that not only induce ferroptosis and cuproptosis, respectively but also lead to cell impairment and eventually diverse cell death. This review summarizes the direct and indirect mechanisms by which Fe and Cu impede cell growth and the various forms of RCD mediated by these two metals. Moreover, we aimed to delineate the interrelationships between these RCDs with the distinct pathways of ferroptosis and cuproptosis, shedding light on the complex and intricate mechanisms that govern cellular survival and death. Finally, the prospects outlined in this review suggest a novel approach for investigating cell death, which may involve integrating current therapeutic strategies and offer a promising solution to overcome drug resistance in certain diseases. Video Abstract.


Asunto(s)
Ferroptosis , Muerte Celular Regulada , Muerte Celular , Cobre , Hierro , Apoptosis
4.
Acta Haematol ; 145(1): 9-17, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34515042

RESUMEN

Immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by persistent thrombocytopenia resulting from increased platelet destruction and a loss of autoimmune tolerance. The pathogenesis of ITP is highly complex. Although ITP may be effectively controlled with currently available medications in some patients, a subset of cases remain refractory. The application of mesenchymal stem cells (MSCs) for human hematopoietic stem cell transplantation has increasingly demonstrated that MSCs modulate innate or adaptive immunity, thus resulting in a tolerant microenvironment. Functional defects and immunomodulatory disorders have been observed after the use of bone marrow mesenchymal stem cells (BM-MSCs) from patients with ITP. Here, we summarize the underlying mechanisms and clinical applications of various derived MSCs for ITP treatment, focusing on the main mechanisms underlying the functional defects and immune dysfunction of BM-MSCs from patients with ITP. Functional effects associated with the activation of the p53 pathway include decreased activity of the phosphatidylinositol 3 kinase/Akt pathway and activation of the TNFAIP3/NF-κB/SMAD7 pathway. Immune dysfunction appears to be associated with an impaired ability of BM-MSCs to induce various types of immune cells in ITP. At present, research focusing on MSCs in ITP remains in preliminary stages. The application of autologous or exogenous MSCs in the clinical treatment of ITP has been attempted in only a small case study and must be validated in larger-scale clinical trials.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas/inmunología , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/terapia , Transducción de Señal/inmunología , Humanos , FN-kappa B/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Proteína p53 Supresora de Tumor/inmunología
5.
Proc Natl Acad Sci U S A ; 116(41): 20528-20538, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548378

RESUMEN

SOX2 is a key transcription factor that plays critical roles in maintaining stem cell property and conferring drug resistance. However, the underlying mechanisms by which SOX2 level is precisely regulated remain elusive. Here we report that MLN4924, also known as pevonedistat, a small-molecule inhibitor of neddylation currently in phase II clinical trials, down-regulates SOX2 expression via causing accumulation of MSX2, a known transcription repressor of SOX2 expression. Mechanistic characterization revealed that MSX2 is a substrate of FBXW2 E3 ligase. FBXW2 binds to MSX2 and promotes MSX2 ubiquitylation and degradation. Likewise, FBXW2 overexpression shortens the protein half-life of MSX2, whereas FBXW2 knockdown extends it. We further identified hypoxia as a stress condition that induces VRK2 kinase to facilitate MSX2-FBXW2 binding and FBXW2-mediated MSX2 ubiquitylation and degradation, leading to SOX2 induction via derepression. Biologically, expression of FBXW2 or SOX2 promotes tumor sphere formation, which is blocked by MSX2 expression. By down-regulating SOX2 through inactivation of FBXW2 E3 ligase, MLN4924 sensitizes breast cancer cells to tamoxifen in both in vitro and in vivo cancer cell models. Thus, a negative cascade of the FBXW2-MSX2-SOX2 axis was established, which regulates stem cell property and drug resistance. Finally, an inverse correlation of expression was found between FBXW2 and MSX2 in lung and breast cancer tissues. Collectively, our study revealed an anticancer mechanism of MLN4924. By inactivating FBXW2, MLN4924 caused MSX2 accumulation to repress SOX2 expression, leading to suppression of stem cell property and sensitization of breast cancer cells to tamoxifen.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas F-Box/metabolismo , Proteínas F-Box/fisiología , Proteínas de Homeodominio/metabolismo , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Factores de Transcripción SOXB1/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Apoptosis , Proliferación Celular , Ciclopentanos/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas F-Box/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Pronóstico , Pirimidinas/farmacología , Factores de Transcripción SOXB1/genética , Tasa de Supervivencia , Tamoxifeno/farmacología , Células Tumorales Cultivadas , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biol Chem ; 295(18): 5906-5917, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32165494

RESUMEN

We previously reported that overexpression of cytochrome P450 family 24 subfamily A member 1 (CYP24A1) increases lung cancer cell proliferation by activating RAS signaling and that CYP24A1 knockdown inhibits tumor growth. However, the mechanism of CYP24A1-mediated cancer cell proliferation remains unclear. Here, we conducted cell synchronization and biochemical experiments in lung adenocarcinoma cells, revealing a link between CYP24A1 and anaphase-promoting complex (APC), a key cell cycle regulator. We demonstrate that CYP24A1 expression is cell cycle-dependent; it was higher in the G2-M phase and diminished upon G1 entry. CYP24A1 has a functional destruction box (D-box) motif that allows binding with two APC adaptors, CDC20-homologue 1 (CDH1) and cell division cycle 20 (CDC20). Unlike other APC substrates, however, CYP24A1 acted as a pseudo-substrate, inhibiting CDH1 activity and promoting mitotic progression. Conversely, overexpression of a CYP24A1 D-box mutant compromised CDH1 binding, allowing CDH1 hyperactivation, thereby hastening degradation of its substrates cyclin B1 and CDC20, and accumulation of the CDC20 substrate p21, prolonging mitotic exit. These activities also occurred with a CYP24A1 isoform 2 lacking the catalytic cysteine (Cys-462), suggesting that CYP24A1's oncogenic potential is independent of its catalytic activity. CYP24A1 degradation reduced clonogenic survival of mutant KRAS-driven lung cancer cells, and calcitriol treatment increased CYP24A1 levels and tumor burden in Lsl-KRASG12D mice. These results disclose a catalytic activity-independent growth-promoting role of CYP24A1 in mutant KRAS-driven lung cancer. This suggests that CYP24A1 could be therapeutically targeted in lung cancers in which its expression is high.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Biocatálisis , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Vitamina D3 24-Hidroxilasa/metabolismo , Adenocarcinoma del Pulmón/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Regulación hacia Arriba , Vitamina D3 24-Hidroxilasa/genética
7.
Int J Exp Pathol ; 102(3): 157-162, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33993564

RESUMEN

Xeroderma Pigmentosum group D (XPD) gene has been shown to suppress hepatocellular carcinoma (HCC) progression, but its mechanism remains not fully understood. ETS-related gene (ERG) is generally known as an oncogenic gene. This study aimed to explore whether XPD regulated HCC cell proliferation, apoptosis and cell cycle by inhibiting ERG expression via the PPARγ pathway. The human hepatoma cells (HepG2) were transfected with the XPD overexpression vector (pEGFP-N2/XPD) or empty vector (pEGFP-N2). The PPARγ inhibitor GW9662 was used to determine whether XPD effects were mediated by activation of PPARγ pathway. Cell cycle and apoptosis were ascertained by flow cytometry, and cell viability was measured by MTT assay. Reverse transcription-polymerase chain reaction and Western blot were performed to determine the mRNA and protein levels. Overexpression of XPD significantly enhanced the expression of PPARγ and p-PPARγ, whereas it downregulated that of ERG and cdk7. Furthermore, XPD overexpression notably inhibited proliferation, promoted apoptosis and decreased the percentage of cells in the S + G2 phase of HepG2 cells. However, these effects of XPD overexpression were abrogated by GW9662. Collectively, XPD suppresses proliferation and promotes apoptosis of HepG2 cells by downregulating ERG expression via activation of the PPARγ pathway.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , PPAR gamma/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo , Apoptosis/fisiología , Carcinoma Hepatocelular/metabolismo , Proliferación Celular/fisiología , Regulación hacia Abajo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Transducción de Señal/fisiología , Regulador Transcripcional ERG/metabolismo
8.
Genes Dev ; 27(14): 1557-67, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23873940

RESUMEN

Metastasis accounts for the vast majority of cancer-related deaths, yet the molecular mechanisms that drive metastatic spread remain poorly understood. Here we report that Tks5, which has been linked to the formation of proteolytic cellular protrusions known as invadopodia, undergoes an isoform switch during metastatic progression in a genetically engineered mouse model of lung adenocarcinoma. Nonmetastatic primary tumor-derived cells predominantly expressed a short isoform, Tks5short, while metastatic primary tumor- and metastasis-derived cells acquired increased expression of the full-length isoform Tks5long. This elevation of Tks5long to Tks5short ratio correlated with a commensurate increase in invadopodia activity in metastatic cells compared with nonmetastatic cells. Further characterization of these isoforms by knockdown and overexpression experiments demonstrated that Tks5long promoted invadopodia in vitro and increased metastasis in transplant models and an autochthonous model of lung adenocarcinoma. Conversely, Tks5short decreased invadopodia stability and proteolysis, acting as a natural dominant-negative inhibitor to Tks5long. Importantly, high Tks5long and low Tks5short expressions in human lung adenocarcinomas correlated with metastatic disease and predicted worse survival of early stage patients. These data indicate that tipping the Tks5 isoform balance to a high Tks5long to Tks5short ratio promotes invadopodia-mediated invasion and metastasis.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Fosfoproteínas/genética , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Animales , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteínas de Unión a Fosfato , Isoformas de Proteínas , Análisis de Supervivencia
9.
Transfusion ; 60(8): 1765-1772, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32627216

RESUMEN

BACKGROUND: The lack of effective treatments against the 2019 coronavirus disease (COVID-19) has led to the exploratory use of convalescent plasma for treating COVID-19. Case reports and case series have shown encouraging results. This study investigated SARS-CoV-2 antibodies and epidemiological characteristics in convalescent plasma donors, to identify criteria for donor selection. METHODS: Recovered COVID-19 patients, aged 18-55 years, who had experienced no symptoms for more than 2 weeks, were recruited. Donor characteristics such as disease presentations were collected and SARS-CoV-2 N-specific IgM, IgG, and S-RBD-specific IgG levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: Whereas levels of N-specific IgM antibody declined after recovery, S-RBD-specific and N-specific IgG antibodies increased after 4 weeks from the onset of symptoms, with no significant correlation to age, sex, or ABO blood type. Donors with the disease presentation of fever exceeding 38.5°C or lasting longer than 3 days exhibited higher levels of S-RBD-specific IgG antibodies at the time of donation. Of the 49 convalescent plasma donors, 90% had an S-RBD-specific IgG titer of ≥1:160 and 78% had a titer of ≥1:640 at the time of plasma donation. Of the 30 convalescent plasma donors, who had donated plasma later than 28 days after the onset of symptoms and had a disease presentation of fever lasting longer than 3 days or a body temperature exceeding 38.5°C, 100% had an S-RBD-specific IgG titer of ≥1:160 and 93% had a titer of ≥1:640. CONCLUSION: This study indicates that the S-RBD-specific IgG antibody reaches higher levels after 4 weeks from the onset of COVID-19 symptoms. We recommend the following selection criteria for optimal donation of COVID-19 convalescent plasma: 28 days after the onset of symptoms and with a disease presentation of fever lasting longer than 3 days or a body temperature exceeding 38.5°C. Selection based on these criteria can ensure a high likelihood of achieving sufficiently high S-RBD-specific IgG titers.


Asunto(s)
Anticuerpos Antivirales/sangre , Betacoronavirus/inmunología , Donantes de Sangre , Convalecencia , Infecciones por Coronavirus/sangre , Neumonía Viral/sangre , Adolescente , Adulto , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/terapia , Fiebre , Humanos , Inmunización Pasiva/normas , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Persona de Mediana Edad , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/patología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología , Factores de Tiempo , Sueroterapia para COVID-19
10.
Cell Biol Toxicol ; 36(4): 349-364, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31907687

RESUMEN

Protein neddylation, a process of conjugating neural precursor cell expressed, developmentally downregulated 8 (NEDD8) to substrates, plays a tumor-promoting role in lung carcinogenesis. Our previous study showed MLN4924, an inhibitor of NEDD8 activating enzyme (E1), significantly inhibits the growth of multiple cancer cells. However, resistance can develop to MLN4924 by mutation. Therefore, it is important to further understand how NEDD8 acts in lung cancer. In the present study, we demonstrated NEDD8 is overactivated in lung cancers and confers a worse patient overall survival. Furthermore, we report that in lung adenocarcinoma cells, NEDD8 depletion significantly suppressed lung cancer cell growth and progression both in vitro and in vivo. Mechanistic studies revealed that NEDD8 depletion induced the accumulation of a panel of tumor-suppressive cullin-RING ubiquitin ligase substrates (e.g., p21, p27, and Wee1) via blocking their degradation, triggering cell cycle arrest at G2 phase, thus inducing apoptosis or senescence in a cell-line-dependent manner. The present study demonstrates the role of NEDD8 in regulating the malignant phenotypes of lung cancer cells and further validates NEDD8 as a potential therapeutic target in lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Proteína NEDD8/metabolismo , Adenocarcinoma del Pulmón/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclopentanos/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo
11.
Genome Res ; 25(7): 1068-79, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26063736

RESUMEN

High-throughput RNA sequencing has revealed more pervasive transcription of the human genome than previously anticipated. However, the extent of natural antisense transcripts' (NATs) expression, their regulation of cognate sense genes, and the role of NATs in cancer remain poorly understood. Here, we use strand-specific paired-end RNA sequencing (ssRNA-seq) data from 376 cancer samples covering nine tissue types to comprehensively characterize the landscape of antisense expression. We found consistent antisense expression in at least 38% of annotated transcripts, which in general is positively correlated with sense gene expression. Investigation of sense/antisense pair expressions across tissue types revealed lineage-specific, ubiquitous and cancer-specific antisense loci transcription. Comparisons between tumor and normal samples identified both concordant (same direction) and discordant (opposite direction) sense/antisense expression patterns. Finally, we provide OncoNAT, a catalog of cancer-related genes with significant antisense transcription, which will enable future investigations of sense/antisense regulation in cancer. Using OncoNAT we identified several functional NATs, including NKX2-1-AS1 that regulates the NKX2-1 oncogene and cell proliferation in lung cancer cells. Overall, this study provides a comprehensive account of NATs and supports a role for NATs' regulation of tumor suppressors and oncogenes in cancer biology.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , ARN sin Sentido/genética , Transcriptoma , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Sitios Genéticos , Humanos , Especificidad de Órganos/genética
12.
Transfusion ; 58(10): 2388-2394, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30203553

RESUMEN

BACKGROUND: Although platelet microparticles (PMPs) were the most abundant micoparticles (MPs) in platelet (PLT) products, other MPs and their parental cells can also be brought into the plasma during PLT apheresis. However, the effects of PLT apheresis, storage time, and leukofiltration on these MPs remain largely unclear. STUDY DESIGN AND METHODS: Apheresis PLTs with or without leukofiltration were stored in 22 ± 2 °C for 5 days. PLT-poor plasma (PPP) was generated by centrifugation of donor blood or PLT products at 2500 × g for 15 minutes on the point day. PPP was labeled with CD41a (PLT-derived MP, PMP), CD235 (red blood cell-derived MP, RMP), CD45 (leukocyte-derived MP, LMP), CD14 (monocyte-derived MP, MMP), and CD144 (endothelial cell-derived MP, EMP), and then measured by flow cytometry. RESULTS: Higher-level TMPs (total microparticles) and PMPs, but lower-level RMPs, LMPs, and MMPs were detected in fresh PLTs on the day of collection compared with those before collection. During storage, TMP, PMP, and RMP counts were significantly higher on Day 3 and Day 5, but MMP and LMP counts were only marginally higher on Day 3 in PLT supernatants. There were no significant differences in MP levels in PLTs with or without leukofiltration. CONCLUSION: MP formation was affected by the apheresis procedure. RMPs, LMPs and MMPs were lower after PLT apheresis. During storage, TMPs, PMPs, RMPs, LMPs, and MMPs were found to be higher in PLT supernatants. Leukofiltration exerted no significant effect on all MPs in PLT products.


Asunto(s)
Conservación de la Sangre/métodos , Micropartículas Derivadas de Células , Procedimientos de Reducción del Leucocitos/métodos , Plaquetoferesis/normas , Plaquetas/citología , Humanos
13.
Wei Sheng Wu Xue Bao ; 57(1): 87-96, 2017 Jan 04.
Artículo en Zh | MEDLINE | ID: mdl-29746763

RESUMEN

Objective: In this study, we constructed recombinant Kluyveromyces lactis strains to produce phospholipase C (PLC) of Bacillus cereus. The recombinant enzymes were purified and characterized. Methods: We cloned the PLC encoding gene bcplc of Bacillus cereus. And the amplified fragments were inserted into pKLAC1 to obtain expression plasmids. K. lactis harboring the above plasmids was cultivated to express PLC that was purified by HisTrapTM affinity chromatography and characterized. Results: PLC of B. cereus was cloned and expressed in K. lactis. The recombinant enzyme had shown activity of 19251 U/mg when using p-nitrophenyl phosphorycholine as substrate. Purified PLC exhibited optimum temperature at 80 °C and optimal pH at 9.0. The recombinant enzyme was stable below 40 °C and pH between 7.0 and 8.0. Cu2+ and Co2+ inhibited its activity whereas Zn2+, Mn2+, Ca2+ and Mg2+ stimulated its activity. Conclusion: It is the first time to express and characterize the PLC gene in K. lactis. These research results provide reference for the study of recombinant PLC.


Asunto(s)
Bacillus cereus/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Expresión Génica , Kluyveromyces/genética , Fosfolipasas de Tipo C/química , Fosfolipasas de Tipo C/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Clonación Molecular , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Kluyveromyces/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Temperatura , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo
14.
Tumour Biol ; 37(5): 6027-34, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26596840

RESUMEN

Pentose phosphate pathway (PPP) is a metabolic pathway that generates NADPH and pentose. PPP genes have been reported to be primarily or secondarily upregulated in many cancers. We aimed to study the general alteration of PPP in acute myelogenous leukemia (AML). We performed data mining and analysis of the Cancer Genome Atlas (TCGA) AML dataset for genetic alteration of the PPP gene set. In vitro studies including proliferation, migration, and invasion assays, together with metabolite consumption and oxidation assays, were performed. PPP genes were upregulated in 61 % of patients with AML. The majority of altered cases were expression changes measured by RNA sequencing. Expressions of critical PPP genes such as G6PD, PFKL, PFKP, and PGLS were consistently upregulated in all altered cases. Altered PPP is not associated with survival or disease relapse. PPP inhibition using 6-aminonicotinamide (6AN) increases glucose oxidative metabolism in AML. 6AN decreased the glucose oxidation and increased fatty acid oxidation. Here, we showed that PPP inhibition increased glucose oxidative metabolism in AML. PPP inhibition suppressed growth, migration, and invasion of AML, but not colony formation. PPP plays an important role in AML. Our results could contribute to the development of novel targeted treatment.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Vía de Pentosa Fosfato , 6-Aminonicotinamida/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Variación Genética , Glucosa/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Oxidación-Reducción/efectos de los fármacos , Pronóstico
15.
Biometrics ; 72(2): 554-62, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26676324

RESUMEN

The discriminatory ability of a marker for censored survival data is routinely assessed by the time-dependent ROC curve and the c-index. The time-dependent ROC curve evaluates the ability of a biomarker to predict whether a patient lives past a particular time t. The c-index measures the global concordance of the marker and the survival time regardless of the time point. We propose a Bayesian semiparametric approach to estimate these two measures. The proposed estimators are based on the conditional distribution of the survival time given the biomarker and the empirical biomarker distribution. The conditional distribution is estimated by a linear-dependent Dirichlet process mixture model. The resulting ROC curve is smooth as it is estimated by a mixture of parametric functions. The proposed c-index estimator is shown to be more efficient than the commonly used Harrell's c-index since it uses all pairs of data rather than only informative pairs. The proposed estimators are evaluated through simulations and illustrated using a lung cancer dataset.


Asunto(s)
Teorema de Bayes , Análisis Discriminante , Modelos Estadísticos , Análisis de Supervivencia , Biomarcadores , Simulación por Computador , Interpretación Estadística de Datos , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Curva ROC , Factores Sexuales
16.
Cancer Cell ; 12(5): 419-31, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17996646

RESUMEN

The Polycomb group (PcG) protein EZH2 possesses oncogenic properties for which the underlying mechanism is unclear. We integrated in vitro cell line, in vivo tumor profiling, and genome-wide location data to nominate key targets of EZH2. One of the candidates identified was ADRB2 (Adrenergic Receptor, Beta-2), a critical mediator of beta-adrenergic signaling. EZH2 is recruited to the ADRB2 promoter and represses ADRB2 expression. ADRB2 inhibition confers cell invasion and transforms benign prostate epithelial cells, whereas ADRB2 overexpression counteracts EZH2-mediated oncogenesis. ADRB2 is underexpressed in metastatic prostate cancer, and clinically localized tumors that express lower levels of ADRB2 exhibit a poor prognosis. Taken together, we demonstrate the power of integrating multiple diverse genomic data to decipher targets of disease-related genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Genómica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Adrenérgicos beta 2/fisiología , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Trasplante de Neoplasias , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Receptores Adrenérgicos beta 2/metabolismo
17.
Exp Cell Res ; 326(1): 78-89, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24880126

RESUMEN

Focal adhesion kinase (FAK) acts as a regulator of cellular signaling and may promote cell spreading, motility, invasion and survival in malignancy. Elevated expression and activity of FAK frequently correlate with tumor cell metastasis and poor prognosis in breast cancer. However, the mechanisms by which the turnover of FAK is regulated remain elusive. Here we report that heat shock protein 90ß (HSP90ß) interacts with FAK and the middle domain (amino acids 233-620) of HSP90ß is mainly responsible for this interaction. Furthermore, we found that HSP90ß regulates FAK stability since HSP90ß inhibitor 17-AAG triggers FAK ubiquitylation and subsequent proteasome-dependent degradation. Moreover, disrupted FAK-HSP90ß interaction induced by 17-AAG contributes to attenuation of tumor cell growth, migration, and invasion. Together, our results reveal how HSP90ß regulates FAK stability and identifies a potential therapeutic strategy to breast cancer.


Asunto(s)
Movimiento Celular , Quinasa 1 de Adhesión Focal/metabolismo , Glicoproteínas de Membrana/metabolismo , Ubiquitina/metabolismo , Apoptosis , Western Blotting , Adhesión Celular , Proliferación Celular , Citoesqueleto/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Invasividad Neoplásica , Fosforilación , Proteolisis , ARN Interferente Pequeño/genética , Transducción de Señal , Células Tumorales Cultivadas , Ubiquitinación
18.
Carcinogenesis ; 35(6): 1292-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24510113

RESUMEN

In cancer cells, the process of epithelial-mesenchymal transition (EMT) confers migratory and invasive capacity, resistance to apoptosis, drug resistance, evasion of host immune surveillance and tumor stem cell traits. Cells undergoing EMT may represent tumor cells with metastatic potential. Characterizing the EMT secretome may identify biomarkers to monitor EMT in tumor progression and provide a prognostic signature to predict patient survival. Utilizing a transforming growth factor-ß-induced cell culture model of EMT, we quantitatively profiled differentially secreted proteins, by GeLC-tandem mass spectrometry. Integrating with the corresponding transcriptome, we derived an EMT-associated secretory phenotype (EASP) comprising of proteins that were differentially upregulated both at protein and mRNA levels. Four independent primary tumor-derived gene expression data sets of lung cancers were used for survival analysis by the random survival forests (RSF) method. Analysis of 97-gene EASP expression in human lung adenocarcinoma tumors revealed strong positive correlations with lymph node metastasis, advanced tumor stage and histological grade. RSF analysis built on a training set (n = 442), including age, sex and stage as variables, stratified three independent lung cancer data sets into low-, medium- and high-risk groups with significant differences in overall survival. We further refined EASP to a 20 gene signature (rEASP) based on variable importance scores from RSF analysis. Similar to EASP, rEASP predicted survival of both adenocarcinoma and squamous carcinoma patients. More importantly, it predicted survival in the early-stage cancers. These results demonstrate that integrative analysis of the critical biological process of EMT provides mechanism-based and clinically relevant biomarkers with significant prognostic value.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Fenotipo , Adulto , Anciano , Línea Celular Tumoral , Análisis por Conglomerados , Biología Computacional , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Proteómica
19.
Eur J Haematol ; 93(4): 329-39, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24766194

RESUMEN

OBJECTIVES: Aspirin (ASA) has been frequently used for thromboprophylaxis in patients with multiple myeloma (MM) when treated with thalidomide or lenalidomide. Despite the well-recognized chemopreventive role of ASA in some solid tumors particularly for colon cancer, whether ASA displays the antimyeloma activity remains unclear. METHODS: MM1.S and RPMI-8226 cell lines harboring K-Ras and N-Ras mutation, respectively, were treated with various concentrations of ASA for different hours. The cell proliferation and apoptosis were performed to explore the effects of ASA on myeloma. Then, the exact mechanisms governing ASA's antimyeloma were explored by qRT-PCR and Western blot. Also, the effect of ASA on tumor growth was observed in NOD/SCID mice bearing myeloma xenografts. RESULTS: ASA of 0-10 mm concentration inhibits proliferation MM1.S and RPMI-8226 cells in time- and dose-dependent manner. The myeloma cells exposed to ASA treatment displayed concentration-dependent apoptosis, which was closely associated with activation of caspases, upregulation of Bax, and downregulation of Bcl-2 and VEGF. Study in vivo revealed that ASA administration retarded the tumor growth accompanying the survival time of mice bearing myeloma xenografts. CONCLUSIONS: ASA exerted antiproliferative and pro-apoptotic action in myeloma cells in vitro and delayed the growth of human myeloma cells in vivo. The underlying mechanisms were ascribed to regulation of Bcl-2 and Bax and suppression of VEGF.


Asunto(s)
Apoptosis/efectos de los fármacos , Aspirina/farmacología , Mieloma Múltiple/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Aspirina/administración & dosificación , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/genética
20.
J Surg Res ; 187(1): 6-13, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24418519

RESUMEN

BACKGROUND: When presenting with advanced stage disease, lung cancer patients have <5% 5-y survival. The overexpression of checkpoint kinase 1 (CHK1) is associated with poorer outcomes and may contribute to therapy resistance. Targeting CHK1 with small-molecule inhibitors in p53 mutant tumors might improve the effectiveness of chemotherapy and radiotherapy in non-small cell lung cancer (NSCLC). METHODS: We evaluated CHK1 messenger RNA and protein levels in multiple NSCLC cell lines. We assessed cell line sensitization to gemcitabine, pemetrexed, and radiotherapy by CHK1 inhibition with the small molecule AZD7762 using proliferation and clonogenic cell survival assays. We analyzed CHK1 signaling by Western blotting to confirm that AZD7762 inhibits CHK1. RESULTS: We selected two p53 mutant NSCLC cell lines with either high (H1299) or low (H1993) CHK1 levels for further analysis. We found that AZD7762 sensitized both cell lines to gemcitabine, pemetrexed, and radiotherapy. Chemosensitization levels were greater, however, for the higher CHK1 protein expressing cell line, H1299, when compared with H1993. Furthermore, analysis of the CHK1 signaling pathway showed that H1299 cells have an increased dependence on the CHK1 pathway in response to chemotherapy. There was no increased sensitization to radiation in H1299 versus H1993. CONCLUSIONS: CHK1 inhibition by AZD7762 preferentially sensitizes high CHK1 expressing cells, H1299, to anti-metabolite chemotherapy as compared with low CHK1 expressing H1993 cells. Thus, CHK1 inhibitors may improve the efficacy of standard lung cancer therapies, especially for those subgroups of tumors harboring higher expression levels of CHK1 protein.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteínas Quinasas/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos/genética , Glutamatos/uso terapéutico , Guanina/análogos & derivados , Guanina/uso terapéutico , Humanos , Neoplasias Pulmonares/metabolismo , Pemetrexed , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/metabolismo , Tiofenos/uso terapéutico , Urea/análogos & derivados , Urea/uso terapéutico , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA