Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Histol Histopathol ; : 18742, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38634557

RESUMEN

A model construction of systemic acute leukemia is challenging. Herein, we established a systemic leukemia mouse model using highly immunodeficient NPG mice without any immunosuppressive treatments. NPG mice received tail intravenous injection of SHI-1 cells at the concentration of 1×107 cells (group A) or 5×107 cells (group B) and randomly sacrificed each seven days post-inoculation. Tumor development was monitored using nested-PCR, peripheral blood-smear analysis, flow cytometry, pathological examinations, and immunohistochemistry. The median survival of mice in groups A and B were 33.0 and 30.0 days, respectively. Blast cells in peripheral blood appeared on day 14 in group B, and on day 21 in group A. In addition, SHI-1 cell specific MLL-AF6 mRNA was detected in both spleen and bone marrow on day 14 post-inoculation. 21 days after inoculation, we observed human CD45+CD33+ cells with an SH-1-immunophenotype in the peripheral blood, spleen, and bone marrow, as well as solid neoplasms in multiple organs. Moreover, the histologically infiltrated leukemic cells expressed CD45. In conclusion, the current study demonstrated the normal growth of SHI-1 cells in the NPG mice without immunosuppression, which caused systemic leukemia similar to that observed in acute leukemia patients. We developed an efficient and reproducible model to study leukemia pathogenesis and progression.

2.
Int J Hematol ; 119(5): 541-551, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38530586

RESUMEN

This study investigated the effect of rapamycin alone and in combination with chemotherapy (doxorubicin and cytarabine) on AML. Human acute monocytic leukemia cell line SHI-1 and NPG AML model mice created by intravenous injection of SHI-1 cell were treated with rapamycin, chemotherapy, or rapamycin plus chemotherapy. Analysis by cell counting kit-8, western blot, flow cytometry, and immunohistochemistry was performed, and results suggested that both rapamycin and chemotherapy inhibited proliferation of SHI-1 cells both in vitro and in vivo, suppressed neoplasm growth in vivo, and promoted survival of NPG AML mice. The antitumor effect of rapamycin plus chemotherapy was better than that of rapamycin alone and chemotherapy alone. In addition, western blot results demonstrated that rapamycin inhibited the phosphorylation of mTOR downstream targets 4EBP1 and S6K1 in SHI-1 cells, and increased the pro-apoptosis-related protein Bax and autophagy-associated proteins Beclin-1, LC3B-II, and ATG5 while reducing the anti-apoptosis-related protein Bcl-2. In conclusion, the results of this study indicate that rapamycin acts synergistically with doxorubicin and cytarabine in AML treatment, and its underlying mechanism might be associated with mTORC1 pathway-mediated apoptosis and autophagy.


Asunto(s)
Apoptosis , Autofagia , Doxorrubicina , Diana Mecanicista del Complejo 1 de la Rapamicina , Transducción de Señal , Sirolimus , Animales , Autofagia/efectos de los fármacos , Apoptosis/efectos de los fármacos , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Sirolimus/farmacología , Línea Celular Tumoral , Doxorrubicina/farmacología , Transducción de Señal/efectos de los fármacos , Citarabina/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Sinergismo Farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Proliferación Celular/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
3.
Small ; : e2308860, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168096

RESUMEN

Developing a new strategy to retain phosphoric acid (PA) to improve the performance and durability of high-temperature proton exchange membrane fuel cell (HT-PEMFC) remains a challenge. Here, a strategy for ion-restricted catcher microstructure that incorporates PA-doped multi-quaternized poly(fluorene alkylene-co-biphenyl alkylene) (PFBA) bearing confined nanochannels is reported. Dynamic analysis reveals strong interaction between side chains and PA molecules, confirming that the microstructure can improve PA retention. The PFBA linked with triquaternary ammonium side chain (PFBA-tQA) shows the highest PA retention rate of 95%. Its H2 /O2 fuel cell operates within 0.6% voltage decay at 160 °C/0% RH, and it also runs over 100 h at 100 °C/49% RH under external humidification. This combination of high PA retention, and chemical and dimensional stability fills a gap in the HT-PEMFC field, which requires strict moisture control at 90-120 °C to prevent acid leaching, simplifying the start-up procedure of HT-PEMFC without preheating.

4.
Small ; 20(4): e2305613, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37712119

RESUMEN

Covalent organic frameworks (COFs) are a family of engaging membrane materials for molecular separation, which remain challenging to fabricate in the form of thin-film composite membranes due to slow crystal growth and insoluble powder. Here, an additive approach is presented to construct COF-based thin-film composite membranes in 10 min via COF oligomer coating onto poly(ether ether ketone) (PEEK)ultrafiltration membranes. By the virtue of ultra-thin liquid phase and liquid-solid interface-confined assembly, the COF oligomers are fast stacked up and grow along the interface with the solvent evaporation. Benefiting from the low out-plane resistance of COFs, COF@PEEK composite membranes exhibit high solvent permeances in a negative correlation with solvent viscosity. The well-defined pore structures enable high molecular sieving ability (Mw = 300 g mol-1 ). Besides, the COF@PEEK composite membranes possess excellent mechanical integrities and steadily operate for over 150 h in the condition of high-pressure cross flow. This work not only exemplifies the high-efficiency and scale-up preparation of COF-based thin-film composite membranes but also provides a new strategy for COF membrane processing.

5.
Cancer Med ; 12(15): 16431-16443, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37409640

RESUMEN

BACKGROUND: The aim of this study is to investigate the profiles of gut microbiota and metabolites in acute myelocytic leukemia (AML) patients treated with/without chemotherapy. METHODS: Herein, high-throughput 16S rRNA gene sequencing was performed to analysis gut microbiota profiles, and liquid chromatography and mass spectrometry were performed to analysis metabolites profiles. The correlation between gut microbiota biomarkers identified by LEfSe and differentially expressed metabolites were determined by spearman association analysis. RESULTS: The results showed the distinguished gut microbiota and metabolites profiles between AML patients and control individuals or AML patients treated with chemotherapy. Compared to normal populations, the ratio of Firmicutes to Bacteroidetes was increased at the phylum level than that in AML patients, and LEfSe analysis identified Collinsella and Coriobacteriaceae as biomarkers of AML patients. Differential metabolite analysis indicated that, compared to AML patients, numerous differential amino acids and analogs could be observed in control individuals and AML patients treated with chemotherapy. Interestingly, spearman association analysis demonstrated that plenty of bacteria biomarkers shows statistical correlations with differentially expressed amino acid metabolites. In addition, we found that both Collinsella and Coriobacteriaceae demonstrate remarkable positive correlation with hydroxyprolyl-hydroxyproline, prolyl-tyrosine, and tyrosyl-proline. CONCLUSION: In conclusion, our present study investigated the role of the gut-microbiome-metabolome axis in AML and revealed the possibility of AML treatment by gut-microbiome-metabolome axis in the further.


Asunto(s)
Microbioma Gastrointestinal , Metabolómica , Humanos , Metabolómica/métodos , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/análisis , Heces/microbiología , Biomarcadores/análisis , Carcinogénesis
6.
ACS Cent Sci ; 9(4): 733-741, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37122458

RESUMEN

High mass transport resistance within the catalyst layer is one of the major factors restricting the performance and low Pt loadings of proton exchange membrane fuel cells (PEMFCs). To resolve the issue, a novel partially ordered phosphonated ionomer (PIM-P) with both an intrinsic microporous structure and proton-conductive functionality was designed as the catalyst binder to improve the mass transport of electrodes. The rigid and contorted structure of PIM-P limits the free movement of the conformation and the efficient packing of polymer chains, resulting in the formation of a robust gas transmission channel. The phosphonated groups provide sites for stable proton conduction. In particular, by incorporating fluorinated and phosphonated groups strategically on the local side chains, an orderly stacking of molecular chains based on group assembly contributes to the construction of efficient mass transport pathways. The peak power density of the membrane electrode assembly with the PIM-P ionomer is 18-379% greater than that of those with commercial or porous catalyst binders at 160 °C under an H2/O2 condition. This study emphasizes the crucial role of ordered structure in the rapid conduction of polymers with intrinsic microporosity and provides a new idea for increasing mass transport at electrodes from the perspective of structural design instead of complex processes.

7.
Int Immunopharmacol ; 119: 110058, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37058751

RESUMEN

BACKGROUND: Accumulating genetic and epigenetic alterations in multiple myeloma (MM) have been demonstrated to be closely associated with osteolytic bone disease, generally characterized as increased osteoclast formation and decreased osteoblast activity. Previously, serum long non-coding RNA (lncRNA) H19 has been proved to be a biomarker for the diagnosis of MM. Whereas, its role in MM-associated bone homeostasis remains largely elusive. METHODS: A cohort of 42 MM patients and 40 healthy volunteers were enrolled for evaluating differential expressions of H19 and its downstream effectors. The proliferative capacity of MM cells was monitored by CCK-8 assay. Alkaline phosphatase (ALP) staining and activity detection, either with Alizarin red staining (ARS) were employed to assess osteoblast formation. Osteoblast- or osteoclast-associated gene were detected using qRT-PCR and western blot analysis. Bioinformatics analysis, RNA pull-down, RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP) were subjected to verify H19/miR-532-3p/E2F7/EZH2 axis, which was accounted for epigenetic suppression of PTEN. The functional role of H19 on MM development through unbalancing osteolysis and osteogenesis was also confirmed in the murine MM model. RESULTS: Upregulation of serum H19 was observed in MM patients, suggesting its positive correlation with the poor prognosis of MM patients. Loss of H19 dramatically weakened cell proliferation of MM cells, promoted osteoblastic differentiation, and impaired osteoclast activity. While reinforced H19 exhibited the opposite effects. Akt/mTOR signaling plays an indispensable role in H19-mediated osteoblast formation and osteoclastgenesis. Mechanistically, H19 served as a sponge for miR-532-3p to upregulate E2F7, a transcriptional activator of EZH2, thereby accounting for modulating epigenetic suppression of PTEN. The in vivo experiments further validated that H19 exerted important impacts on tumor growth through breaking the balance between osteogenesis and osteolysis via Akt/mTOR signaling. CONCLUSION: Collectively, increased enrichment of H19 in MM cells exhibits an essential role in MM development by disturbing bone homeostasis.


Asunto(s)
MicroARNs , Mieloma Múltiple , Osteólisis , ARN Largo no Codificante , Humanos , Ratones , Animales , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Osteogénesis/genética , Proteínas Proto-Oncogénicas c-akt , Mieloma Múltiple/genética , Osteólisis/genética , Diferenciación Celular/genética , Serina-Treonina Quinasas TOR
8.
ACS Appl Mater Interfaces ; 13(49): 58838-58847, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34851081

RESUMEN

The polarization curve is the most important profile to evaluate the performance of proton-exchange membrane fuel cells (PEMFCs). To explore the important thermodynamic parameters and their correlation with the composition, fabrication, and operational settings, a comprehensive data set consisting of 446 polarization curves from 191 perfluorosulfonate and 255 sulfonated hydrocarbon-based PEMs is collected. Then, a Markov chain Monte Carlo simulation within the Bayesian frame provides higher than 93% confidence to extract six important thermodynamic parameters including open-circuit potential, the transfer coefficient, the current loss, the reference exchange current density, the internal resistance, and the limiting current density. An extreme gradient boosting algorithm affords a mean determinative coefficient of 0.89 to predict the whole polarization curve and a confidence of 94% to predict the peak power density (PPD). Both approaches to explore the polarization curve for PEMFCs show good robustness in the blind test. Overall, the PPD is positively correlated with the ion-exchange capacity of the polymer, operational temperature, and humidity and is negatively affected by internal resistance, membrane thickness, and the loading of the catalyst. The flow rate of fuels can effectively enhance them, while the increase of catalyst loading or fuel concentration shows deleterious impacts.

9.
Cell Immunol ; 363: 104342, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33765541

RESUMEN

BACKGROUND: Chimeric antigen receptor T cells (CAR-T) against B-cell maturation antigen (BCMA) has been used to treat multiple myeloma (MM). CAR-T cells co-expressing a truncated human EGFR (tEGFR) has been proposed for in vivo cell ablation. METHODS: We designed and tested a novel anti-BCMA CAR. We transduced T cells with retroviral vectors encoding CAR and tEGFR. The anti-BCMA-CAR-transduced T cells were evaluated for the functions including cytokine production, proliferation, cytotoxicity, and in vivo tumor eradication of BCMA. Cetuximab was used for in vivo cell ablation. RESULTS: The CAR-T cells could specifically recognize BCMA, and anti-BCMA CAR-T cells could exhibit interferon-γ and cytotoxicity specifically produced by BCMA and eradicate tumor in vivo. Cetuximab could mediate antibody-dependent cellular cytotoxicity and in vivo elimination. CONCLUSIONS: We confirm that BCMA is a suitable target for CAR- T cells and tEGFR is a effective tool for cellular ablation.


Asunto(s)
Antígeno de Maduración de Linfocitos B/inmunología , Receptores ErbB/genética , Inmunoterapia Adoptiva/métodos , Adulto , Animales , Antígeno de Maduración de Linfocitos B/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Femenino , Células HEK293 , Xenoinjertos , Humanos , Células K562 , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Pathology ; 53(5): 613-622, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33558065

RESUMEN

LncRNA PVT1 has been demonstrated to be upregulated in acute myeloid leukaemia (AML) patients and indicates a poor prognosis. Nevertheless, its role in AML remains obscure. This study investigated the regulatory role and potential mechanisms of PVT1 in the progression of AML. Expression of PVT1, miR-29 family and WAVE1 was detected by quantitative real-time polymerase chain reaction. CCK8 and EdU assays were performed to assess the proliferation of AML cells. Cell cycle and apoptosis were determined by propidium iodide (PI) staining and Annexin V/PI staining on a flow cytometer. Transwell assay was carried out to evaluate the migration and invasion abilities. The interaction between miR-29 family and PVT1/WAVE1 was confirmed by dual luciferase reporter assay and RNA immunoprecipitation assay. The protein levels of WAVE1, Bcl-2, Bax, cleaved Caspase 3, cyclin D1, and p21 were detected by western blotting. Xenograft transplantation was performed to determine the tumourigenicity of AML cell in vivo. PVT1 expression was significantly increased in AML patient samples and cells, which positively correlated with WAVE1 expression. Silencing of PVT1 restrained growth, migration and invasion, while inducing apoptosis of AML cells. Moreover, PVT1 acted as a sponge for miR-29 family to increase WAVE1 expression in AML cells. Overexpression of WAVE1 partly counteracted PVT1 knockdown-induced anti-tumour effects on AML cells in vitro and xenograft tumour in vivo. PVT1 facilitated the progression of AML via regulating miR-29 family/WAVE1 axis, which supported the conclusion that PVT1 may be a promising therapeutic target for AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos BALB C , Neoplasias/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética
11.
Lab Invest ; 101(3): 341-352, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33318617

RESUMEN

Acute myelogenous leukemia (AML) is the most common acute leukemia in adults. Despite great progress has been made in this field, the pathogenesis of AML is still not fully understood. We report here the biological role of lncRNA small nucleolar RNA host gene 5 (SNHG5) in the pathogenesis of AML and the underlying mechanisms. The results showed that lncRNA SNHG5 was highly expressed in AML cancer cell lines. In vitro studies displayed that inhibition of SNHG5 with shRNA resulted in suppression of survival, cell cycle progression, migration/invasion of AML and capacity of adhesion and angiogenesis in human umbilical vein endothelial cells. Mechanistic studies revealed a SNHG5/miR-26b/connective tissue growth factor (CTGF)/vascular endothelial growth factor A (VEGFA) axis in the regulation of AML angiogenesis. Finally, Yin Yang 1 (YY1) was found to transactivate and interact with SNHG5 promoter, leading to the upregulation of SNHG5 in AML. Collectively, upregulation of lncRNA SNHG5 mediated by YY1, activates CTGF/VEGFA via targeting miR-26b to regulate angiogenesis of AML. Our work provides new insights into the molecular mechanisms of AML.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Neovascularización Patológica/genética , ARN Largo no Codificante/genética , Factor de Transcripción YY1/genética , Adulto , Línea Celular Tumoral , Supervivencia Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Masculino , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción YY1/metabolismo
12.
Chem Commun (Camb) ; 56(80): 12045-12048, 2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-32902526

RESUMEN

A novel liquid-infused, patterned, porous membrane system with anti-fouling characteristics is prepared via simple co-infusion of oil and water within hydrophobic and superhydrophilic surfaces of a porous membrane, respectively. This membrane simultaneously repels the immiscible water and oil exhibiting excellent interfacial floatability at the oil-water interface as a separator, thus showing promise for use in applications in the immiscible oil/water separation industry and liquid-liquid extraction.

13.
Pathol Int ; 70(10): 724-732, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32754978

RESUMEN

Long non-coding RNA (lncRNA) MALAT1 has been confirmed to function as an oncogene in various solid tumors. MALAT1 level has been shown to be upregulated in relapsed acute lymphoblastic leukemia (ALL) patients, but the mechanism is unclear. This study aims to investigate the functional roles and underlying mechanisms of MALAT1 in ALL. MALAT1 and miR-205 expression were assessed by real-time quantitative polymerase chain reaction (RT-qPCR). MTT assay and flow cytometry were performed to evaluate cell proliferation and apoptosis, respectively. Protein level of protein tyrosine kinase-7 (PTK7) was detected by Western blot assay. Dual luciferase reporter assay was conducted to confirm the binding of MALAT1 and miR-205, as well as miR-205 and PTK7. The levels of MALAT1 and PTK7 were upregulated in ALL samples. In contrast, miR-205 level was downregulated in ALL in ALL samples. Moreover, MALAT1 silencing or miR-205 overexpression restrained proliferation and promoted apoptosis of ALL cells. Mechanistically, MALAT1 sponged miR-205 to regulate PTK7 expression. In summary, MALAT1 affected ALL cell proliferation and apoptosis via regulating miR-205-PTK7 axis. Our results suggest that MALAT1-miR-205-PTK7 axis participates in the proliferation and apoptosis of ALL, which may provide a potential treatment target for ALL.


Asunto(s)
Apoptosis/genética , Moléculas de Adhesión Celular/metabolismo , MicroARNs/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , ARN Largo no Codificante/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , ARN Largo no Codificante/genética , Proteínas Tirosina Quinasas Receptoras/genética , Regulación hacia Arriba
14.
Mol Cell Biol ; 40(3)2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31712391

RESUMEN

Multiple myeloma (MM) accounts for over twenty percent of hematological cancer-related death worldwide. Long noncoding RNA (lncRNA) H19 is associated with multiple tumorigenesis and is increased in MM, but the underlying mechanism of H19 in MM is unclear. In this study, the expression of H19, microRNA 152-3p (miR-152-3p), and BRD4 in MM patients was evaluated by quantitative real-time PCR (qRT-PCR) and Western blotting. Colony formation and flow cytometry analysis were used to determine the effects of H19 and miR-152-3p on MM cell proliferation, apoptosis, and cell cycle. A luciferase reporter assay was conducted to confirm the interaction among H19, miR-152-3p, and BRD4. A nude mouse xenograft model was established, and the cell proliferation and apoptosis were evaluated by immunohistochemistry (IHC) staining and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling assay. We found that levels of H19 and BRD4 were upregulated and the expression of miR-152-3p was downregulated in MM patients. Dual luciferase reporter assay showed H19 targeted miR-152-3p to promote BRD4 expression. Knockdown of H19 repressed proliferation and enhanced apoptosis and cell cycle G1 arrest by upregulating miR-152-3p in MM cells. Furthermore, H19 knockdown suppressed the growth of xenograft tumor, reduced Ki-67 and BRD4 levels, and increased cell apoptosis in xenograft tumor tissues. Taking these results together, H19 knockdown suppresses MM tumorigenesis via inhibiting BRD4-mediated cell proliferation through targeting miR-152-3p, implying that H19 is a promising biomarker and drug target for MM.


Asunto(s)
Proteínas de Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Mieloma Múltiple/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Animales , Apoptosis , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Transducción de Señal , Factores de Transcripción/metabolismo
15.
Cancer Med ; 8(17): 7143-7160, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31568697

RESUMEN

BACKGROUND: Recent studies have revealed that long noncoding RNAs (lncRNAs) may hold crucial triggers of the pathogenesis of hematological malignancies, while the studies evaluating the expression pattern of lncRNA in acute myeloid leukemia (AML) are few. Thus, this study aimed to investigate the implication of lncRNA expression pattern in AML development and progression. METHODS: Bone marrow samples from four AML patients and four controls were subjected to lncRNA sequencing. Then, bone marrow samples from 110 AML patients and 40 controls were proposed to real-time quantitative polymerase chain reaction (RT-qPCR) validation for 10 candidate lncRNAs. Clinical data and survival profiles were recorded in AML patients. Furthermore, lncRNA RP4-576H24.2 expression in AML cell lines and its effect on AML cell proliferation and apoptosis were detected. RESULTS: LncRNA expression pattern by sequencing clearly distinguished AML patients from controls, and 630 upregulated and 621 downregulated lncRNAs were identified in AML patients compared to controls, which were mainly enriched in AML oncogene-related biological process and pathways (such as neutrophil degranulation, leukocyte transendothelial migration, and hematopoietic cell lineage). RT-qPCR validation observed that six lncRNAs correlated with AML risk, one lncRNA associated with risk stratification, and three lncRNAs correlated with survivals, among which lncRNA RP4-576H24.2 was the only one correlated with AML susceptibility, risk stratification, and survivals. Further in vitro experiments showed that lncRNA RP4-576H24.2 was upregulated in AML cell lines compared to normal bone marrow mononuclear cells (BMMCs), and promoted proliferation while inhibited apoptosis in HL-60 and KG-1 cells. CONCLUSIONS: LncRNA expression pattern is closely involved in the development and progression of AML, and several specific lncRNAs exhibit potential to be biomarkers for AML risk and prognosis. Besides, lncRNA RP4-576H24.2 might be a potential oncogene in AML pathogenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , ARN Largo no Codificante/metabolismo , Adulto , Biomarcadores de Tumor/análisis , Biopsia , Médula Ósea/patología , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Pronóstico , ARN Largo no Codificante/análisis , Análisis de Secuencia de ARN , Regulación hacia Arriba
16.
J Colloid Interface Sci ; 554: 658-667, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31351336

RESUMEN

Functionalized polysulfone (PSf) membranes with combined antibacterial and antifouling properties were fabricated by incorporating a poly(ethyleneoxide)-grafted (PEO-grafted) amphiliic polymer. Both antifouling and antibacterial groups were easily introduced onto the membrane surfaces through non-solvent induced phase separon process and a simple chlorination process. It was observed that the functionalized membranes were effectivatie in resisting both protein absorption and bacterial adhesion. Furthermore, the functionalized membrane (M3-Cl) showed mostly suppressed irreversible flux decline and a 97% flux recovery ratio after simple washing during the separation process, indicating excellent antifouling properties. Meanwhile, the functionalized PSf membrane exhibited efficient biocidal activity against E. coli and S. aureus. These modified functionalized PSf membranes also displayed outstanding properties in inhibiting the formation of biofilm. Moreover, the antibacterial feature was renewable by a simple process.


Asunto(s)
Antibacterianos/química , Cloraminas/química , Membranas Artificiales , Polietilenglicoles/química , Polímeros/química , Sulfonas/química , Antibacterianos/farmacología , Infecciones Bacterianas/prevención & control , Biopelículas/efectos de los fármacos , Incrustaciones Biológicas/prevención & control , Cloraminas/farmacología , Escherichia coli/efectos de los fármacos , Humanos , Polietilenglicoles/farmacología , Polímeros/farmacología , Staphylococcus aureus/efectos de los fármacos , Sulfonas/farmacología
17.
Biosci Rep ; 39(5)2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30455393

RESUMEN

Background: Multiple myeloma (MM) is an incurable hematologic cancer, accompanied by excessive osteoclast formation and inflammatory cytokine secretion. The mechanisms by which bromodomain and extra-terminal domain (BET) protein inhibitor I-BET151 regulates osteoclast differentiation and inflammatory cytokine secretion in MM are largely unknown. Methods: The isolated peripheral blood mononuclear cells from normal or patients with MM were treated with receptor activator of NF-κB ligand (RANKL) and M-CSF to induce osteoclast differentiation. RAW 264.7 cells were treated with RANKL. I-BET151 was applied to investigate the effects of BRD4 inhibition on osteoclast formation and inflammatory cytokine secretion. Osteoclast formation was determined by tartrate-resistant acid phosphatase (TRACP) staining. The expression of osteoclast-specific genes TRACP, matrix metalloproteinase-9 (MMP-9), cathepsin K (Ctsk), and c-Src was tested using quantitative real-time PCR. And the level of inflammatory cytokines TNF-α, IL-1ß, and IL-6 was assessed by ELISA. Tumor necrosis factor receptor-associated factor 6 (TRAF6), BRD4, nuclear and cytoplasm p65, IκB-α, nuclear factor of activated T cells cytoplasmic (NFATc1), and osteoprotegerin (OPG) expression were measured by Western blotting. RNAi technology was applied to knock down BET family member BRD4. Results: I-BET151 dose-dependently suppressed osteoclast formation, inhibited the levels of osteoclast-specific genes TRACP, MMP-9, Ctsk, and c-Src and inflammatory cytokines TNF-α, IL-1ß, and IL-6 secretion in peripheral blood mononuclear cells and RAW 264.7. I-BET151 inhibited the protein levels of BRD4 and NFATc1, increased OPG expression, and suppressed IκB-α degradation and p65 nuclear translocation. Further, the effects of I-BET151 on osteoclast formation, osteoclast-specific genes expression, inflammatory cytokine secretion, and NF-κB inhibition were promoted by BRD4 knockdown. Conclusion: I-BET151 inhibits osteoclast formation and inflammatory cytokine secretion by targetting BRD4-mediated RANKL-NF-κB signal pathway and BRD4 inhibition might be beneficial for MM treatment.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Citocinas/inmunología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Osteogénesis/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/inmunología , Células Cultivadas , Humanos , Ratones , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , FN-kappa B/inmunología , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Osteoclastos/patología , Ligando RANK/inmunología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/inmunología , Células Tumorales Cultivadas
18.
Int J Mol Med ; 42(1): 489-496, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29620147

RESUMEN

Angiogenesis is essential for various biological processes, including tumor blood supply delivery, cancer cell growth, invasion and metastasis. Plasmacytoma variant translocation 1 (PVT1) long noncoding RNA (lncRNA) has been previously reported to affect angiogenesis of glioma microvascular endothelial cells by regulating microRNA (miR)­186 expression level. However, the specific underlying molecular mechanism of PVT1 regulation of angiogenesis in vascular endothelial cells remains to be elucidated. The present study investigated the role of PVT1 in cell proliferation, migration and vascular tube formation of human umbilical vein endothelial cells (HUVECs) using MTT assay, Transwell migration assay and in vitro vascular tube formation assay, respectively. In order to determine the effect of miR­26b on cell proliferation, migration and vascular tube formation of HUVECs, miR­26 mimic or miR­26b inhibitor were transfected into HUVECs. Reverse transcription­quantitative polymerase chain reaction and western blotting were conducted to quantify the mRNA and protein expression levels of target genes. The present study confirmed that miR­26b bound 3'­untranslated region (3'­UTR) and subsequently influenced gene expression level using dual luciferase reporter assay. The current study observed that PVT1 affected cell proliferation, migration and in vitro vascular tube formation of HUVECs. In addition, it was determined that PVT1 was able to bind and degrade miR­26b to promote connective tissue growth factor (CTGF) and angiopoietin 2 (ANGPT2) expression. miR­26b was also identified to have a suppressive role in cell proliferation, migration and in vitro vascular tube formation of HUVECs via binding 3'­UTR regions and downregulating CTGF and ANGPT2 expression levels. The current findings may improve the understanding of the underlying mechanism of PVT1 contributing to angiogenesis of vascular endothelial cells and offer rationale for targeting PVT1 to treat angiogenesis dysfunction­associated diseases, including cancer metastasis.


Asunto(s)
Angiopoyetina 2/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/metabolismo , Neovascularización Fisiológica/genética , ARN Largo no Codificante/metabolismo , Regiones no Traducidas 3'/genética , Secuencia de Bases , Movimiento Celular/genética , Proliferación Celular/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética
19.
Int J Clin Exp Pathol ; 11(1): 68-75, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-31938088

RESUMEN

To investigate the effect of CXCR4 gene expressing on the proliferation, adhesion, and invasion of human monocytic leukemic cell line SHI-1 and its tumorigenic capacity in nude mouse. The SHI-1 cells were firstly infected with the rescued recombinant lentivirus to establish SHI-1/CXCR4i cell line. The expression of CXCR4, MMP-2 and MMP-9 in the SHI-1/CXCR4i cell was determined by real time quantitative PCR and flow cytometry. MTT assay was performed to detect the SHI-1/CXCR4i cell proliferative rate. The co-culture system of the leukemia cells with bone marrow stromal cells was utilized to detect the adhesive and migratory ability of SHI-1/CXCR4i cell. The nude mouse was subcutaneously inoculated with both cell lines, and then used to evaluate the growth ability of leukemia cells without CXCR4 expressing. The expression of CXCR4 mRNA in established SHI-1/CXCR4i cells decreased by 76% comparing with that in SHI-1/NC cells, which were infected with negative control virus. The proliferative rate of the SHI-1/CXCR4i cells in vitro did not show obvious difference with the SHI-1/NC cells, however, the adhesive and trans-Matrigel invasive ability were significantly decreased. Notably, the neoplasm was not observed in mice subcutaneously inoculated with SHI-1/CXCR4i cells, but presented in the SHI-1/WT and SHI-1/NC cells-inoculated mice in neoplastic volume of both groups. The present data showed that the CXCR4 silencing reduced the adhesive and migratory ability of SHI-1 cells in vitro, and suppressed the formation of subcutaneous neoplasm in vivo, demonstrating that the CXCR4 can be served as a novel target for leukemia gene therapy.

20.
Mol Med Rep ; 16(6): 8406-8412, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28983590

RESUMEN

Excessive bone resorption mediated by osteoclasts may lead to the risk of various lytic bone diseases. In the present study, the effects of I­BET151, a bromodomain and extra terminal domain protein inhibitor, on osteoclastogenesis in RAW264.7 cells and the underlying mechanism of this process was investigated. Cells were divided into 6 groups, including the control group, receptor activator of nuclear factor­κB ligand (RANKL) group and 4 other groups containing RANKL and I­BET151 at different concentrations. Tartrate­resistant acid phosphatase (TRACP) staining was used to observe the effect of I­BET151 on osteoclastogenesis and the number of TRACP positive multinucleated cells was calculated. Western blotting was used to evaluate the expression of tumor necrosis factor receptor associated factor (TRAF6), nuclear factor of activated T­cells cytoplasmic 1 (NFATcl), transcription factor p65 (p65), nuclear factor of κ light polypeptide gene enhancer in B­cells inhibitor­α (IκB­α), extracellular signal­regulated kinase, Jun N­terminal kinase (JNK) and p38. mRNA expression levels of osteoclast specific genes TRACP, matrix metalloproteinase­9 (MMP9), cathepsin K (CtsK) and proto­oncogene tyrosine­protein kinase Src (c­Src) were measured using the reverse transcription­quantitative polymerase chain reaction (RT­qPCR). TRACP staining results demonstrated that I­BET151 inhibited osteoclastogenesis induced by RANKL and the inhibition was dose dependent. TRACP multinucleated positive cells were significantly decreased when treated with I­BET151 compared with the RANKL group. The inhibitory effect on TRAF6 was significant when concentrations of 100 and 200 nM I­BET151 were used, and NFATcl was significantly inhibited when a concentration of 200 nM was used compared with the RANKL group, in a dose-dependent manner. Nuclear translocation of p65 was significantly inhibited by I­BET151 at all concentrations. The degradation of IκB­α, and phosphorylation of JNK and p38 were also significantly inhibited by I­BET151, with the exception of the expression of IκB­α following treatment with 50 nM I­BET151. The RT­qPCR results revealed that osteoclast­specific genes TRACP, MMP9, CtsK and c­Src were all dose­dependently inhibited by I­BET151, except for CtsK. In conclusion, I­BET151 may significantly suppress the osteoclastogenesis of RAW264.7 cells via the RANKL signaling pathway.


Asunto(s)
Resorción Ósea/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Resorción Ósea/genética , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...