Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Signal ; 123: 111356, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39173857

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common diagnosed subtype of lymphoma with high invasiveness and heterogeneity. Glycolysis is involved in regulating DLBCL progression. We aimed to explore the role of forkhead box protein A1 (FOXA1) in DLBCL and the mechanisms related to sirtuine5 (SIRT5) and glycolysis. FOXA1 expression in DLBCL cells was analyzed. Then, the proliferation and apoptosis of DLBCL cells were detected using Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EDU) staining and flow cytometry analysis following FOXA1 or SIRT5 knockdown. The glycolysis was assessed by measuring extracellular acidification rate (ECAR), glucose consumption and lactate secretion. Immunoblotting was employed to examine the expression of apoptosis- and glycolysis-related proteins. Additionally, luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to test the combination of FOXA1 to SIRT5 promotor region. Subsequently, SIRT5 expression was upregulated to conduct rescue assays. Finally, the effects of FOXA1 downregulation on the growth and glycolysis in OCI-ly7 tumor-bearing mice were examined. As a result, FOXA1 was upregulated in DLBCL cells and FOXA1 or SIRT5 knockdown inhibited the proliferation, accelerated the apoptosis and suppressed glycolysis reprograming in DLBCL cells. Importantly, FOXA1 could transcriptionally activate SIRT5 expression in DLBCL cells. Besides, SIRT5 overexpression counteracted the effects of FOXA1 deficiency on the proliferation, apoptosis and glycolysis reprogramming in DLBCL cells. Furthermore, FOXA1 knockdown inhibited the tumor growth, suppressed the glycolysis reprogramming and downregulated SIRT5 expression in vivo. In summary, FOXA1 could transcriptionally activate SIRT5 to reprogram glycolysis, thereby facilitating the malignant progression of DLBCL.


Asunto(s)
Glucólisis , Factor Nuclear 3-alfa del Hepatocito , Linfoma de Células B Grandes Difuso , Sirtuinas , Activación Transcripcional , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Animales , Sirtuinas/metabolismo , Sirtuinas/genética , Ratones , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Apoptosis , Progresión de la Enfermedad
2.
Exp Ther Med ; 27(1): 7, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38223325

RESUMEN

Epstein-Barr virus (EBV) has been implicated in the development of a wide range of lymphoproliferative disorders. In this process, the role of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) has remained to be clarified. A meta-analysis of 20 studies was performed and risk ratios (RRs) with 95% confidence intervals (CIs) were used to evaluate the association between PD-L1/PD-1 expression and the status of EBV infection. The results showed that the expression level of PD-L1 in tumor cells was significantly higher in EBV+ cases with a pooled RR of 2.26 (95% CI, 1.63-3.14; P<0.01), particularly in subtypes of diffuse large B-cell lymphoma (DLBCL) and classical Hodgkin lymphoma. Similarly, EBV infection increased the expression of PD-L1 in immune cells with a pooled RR of 2.20 (95% CI, 1.55-3.12; P<0.01). In subtypes of DLBCL and post-transplant lymphoproliferative disorder, the expression of PD-L1 in immune cells is increased in EBV+ cases. Regarding the expression level of PD-1 in tumor-infiltrating lymphocytes (TILs), no significance was found between EBV infection and PD-1 expression, with a pooled RR of 1.10 (95% CI, 0.81-1.48; P>0.05). The present meta-analysis demonstrated that in EBV-associated lymphoproliferative disorders, EBV infection was associated with the expression level of PD-L1 in tumor cells and immune cells but was not associated with the expression of PD-1 in TILs.

4.
Int J Med Sci ; 20(5): 616-626, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082728

RESUMEN

Background: Mantle cell lymphoma (MCL) is a heterogeneous disease belonging to non-Hodgkin's lymphoma. In recent years, the morbidity rate of MCL is ascending, and the prognosis remains unfavorable. Ubiquitin-specific proteases14 (USP14) has been evidenced to be engaged in the process of malignant tumors. In this article, the role of USP14 in the malignant process of MCL and the mechanism of ibrutinib resistance were discussed. Methods: Through qRT-PCR and western blot, the mRNA and protein expressions of USP14 in MCL cells were tested. USP14 interference plasmid was constructed by cell transfection technology, and then CCK8 and EdU assays were applied to appraise cell proliferation. Cell cycle and cell apoptosis were estimated by flow cytometry and western blot. The sensitivity of MCL cells to ibrutinib was also investigated. Next, western blot, co-IP, Cycloheximide (CHX) assay and other techniques were used to detect the relationship between USP14 and XPO1. Finally, by simultaneously inhibiting USP14 and overexpressing XPO1, the impacts of USP14 on the malignant process of MCL and the regulatory mechanism of ibrutinib sensitivity in MCL were discussed. Results: USP14 expression was markedly fortified in MCL cell lines. Interference of USP14 suppressed MCL cell viability, potentiated cell cycle arrest, apoptosis, and ibrutinib sensitivity. This process might be achieved by USP14 deubiquitination through enhancing XPO1 stability. Conclusion: USP14 can promote the malignant progression and ibrutinib sensitivity of MCL by stabilizing XPO1.


Asunto(s)
Linfoma de Células del Manto , Humanos , Adulto , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/metabolismo , Ubiquitina , Línea Celular Tumoral , Piperidinas , Ubiquitina Tiolesterasa/genética
5.
Front Oncol ; 12: 1091782, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36605439

RESUMEN

Immune evasion through up-regulating checkpoint inhibitory receptors on T cells plays an essential role in tumor initiation and progression. Therefore, immunotherapy, including immune checkpoint inhibitor targeting programmed cell death protein 1 (PD-1) and chimeric antigen receptor T cell (CAR-T) therapy, has become a promising strategy for hematological malignancies. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is a novel checkpoint inhibitory receptor expressed on immune cells, including cytotoxic T cells, regulatory T cells, and NK cells. TIGIT participates in immune regulation via binding to its ligand CD155. Blockage of TIGIT has provided evidence of considerable efficacy in solid tumors in preclinical research and clinical trials, especially when combined with PD-1 inhibition. However, the mechanism and function of TIGIT in hematological malignancies have not been comprehensively studied. In this review, we focus on the role of TIGIT in hematological malignancies and discuss therapeutic strategies targeting TIGIT, which may provide a promising immunotherapy target for hematological malignancies.

6.
J Infect ; 80(4): 373-377, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32109444

RESUMEN

A novel coronavirus, designated as 2019-nCoV, hit the central Chinese city of Wuhan in late December 2019, and subsequently spread rapidly to all provinces of China and multiple countries. As of 0:00 am February 9, 2020, a total of 37,287 cases have been confirmed infection of 2019-nCoV in China mainland, and 302 cases have also been cumulatively reported from 24 countries. According to the latest data, a total of 813 deaths occurred in China mainland, with the mortality reaching approximately 2.2%. At present, there is no vaccine or specific drugs for the human coronavirus. Therefore, it is critical to understand the nature of the virus and its clinical characteristics, in order to respond to the 2019-nCoV outbreak. Thus, the present study briefly but comprehensively summarizes the not much but timely reports on the 2019-nCoV.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Pandemias , Neumonía Viral , Factores de Edad , Antivirales/uso terapéutico , Betacoronavirus/química , Betacoronavirus/genética , COVID-19 , China/epidemiología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/virología , Femenino , Genoma Viral , Humanos , Masculino , Neumonía Viral/epidemiología , Neumonía Viral/fisiopatología , Neumonía Viral/terapia , Neumonía Viral/virología , SARS-CoV-2 , Factores Sexuales , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Tratamiento Farmacológico de COVID-19
7.
J Int Med Res ; 46(11): 4418-4426, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30226089

RESUMEN

The function of the immune system in cancer initiation and progression has been widely examined. Notably, immunotherapy has become a promising approach for cancer treatment. CD47, a member of the immunoglobulin superfamily, plays an important role in the immune regulation of cancer by binding to SIRPα. Multiple studies have detected high CD47 expression on the surface of tumor cells, which indicates poor prognosis. Treatments that block the interaction of CD47 and SIRPα significantly suppress tumor growth and metastasis through diverse mechanisms, such as phagocytosis, antibody-dependent cellular cytotoxicity, and apoptosis. Recently, several studies have reported increased CD47 expression on different types of lymphoma cells, indicating that the CD47-SIRPα pathway can be used as a therapeutic target in lymphoma. This review focuses on the role of CD47-SIRPα in B-cell lymphoma and discusses promising therapeutic strategies targeting the CD47-SIRPα axis, which yield insights into the immunotherapy of B-cell lymphoma.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Inmunoterapia , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Receptores Inmunológicos/metabolismo , Transducción de Señal , Ensayos Clínicos como Asunto , Humanos
8.
Oncol Rep ; 39(6): 2473-2481, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29658589

RESUMEN

Autophagy, a physiological process in which cellular components are degraded by the lysosome for cell homeostasis, plays an important role in cell metabolism, including cell proliferation, differentiation, survival and apoptosis. Recent studies indicate that autophagy serves as a survival mechanism by eliminating misfolded proteins and attenuating DNA damage. Autophagy can also suppress tumor growth, depending upon the cell context and functional status. Dysfunction of autophagy may be closely linked to the initiation and development of various diseases, including hematological malignancies. Mounting evidence highlights the dual role of autophagy in blood cancers through multifarious signal pathways. Therefore, strategies targeting autophagy will develop innovative therapeutic approaches for blood cancers, improve the efficacy of chemotherapy, and bring significant benefits for patients.


Asunto(s)
Autofagia , Redes Reguladoras de Genes , Neoplasias Hematológicas/genética , Terapia Molecular Dirigida/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Transducción de Señal
9.
Acta Pharmacol Sin ; 39(2): 251-260, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28905936

RESUMEN

Cancer virotherapy provides a new strategy to treat cancer that can directly kill cancer cells by oncolysis. Insertion of therapeutic genes into the genome of a modified adenovirus, thereby creating a so-called gene-virotherapy that shares the advantages of gene therapy and virotherapy. In this study we investigated whether a strategy that combines the oncolytic effects of an adenoviral vector with the simultaneous expression of the autophagy gene Beclin-1 offered a therapeutic advantage for chronic myeloid leukemia (CML) cells with resistance to chemotherapy and evaluated the synergistic effects of SG511-BECN and doxorubicin (Dox) in human CML cells in vitro. Oncolytic virus SG511-BECN was constructed through introducing the Beclin-1 gene into the oncolytic adenoviral backbone. SG511-BECN displayed significantly improved antileukemia activity on multidrug-resistant CML cell line K562/A02, which was mediated via induction of autophagic cell death. Furthermore, Dox could synergize with SG511-BECN to kill the CML cells by improving the infectious efficiency of the oncolytic adenovirus without causing significant damage to normal human mononuclear cells. The results demonstrate that targeting the autophagic cell death pathway and combination of a chemotherapy agent with oncolytic adenovirus may be a novel strategy for the treatment of leukemia with chemotherapy resistance.


Asunto(s)
Antineoplásicos/farmacología , Beclina-1/metabolismo , Doxorrubicina/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Virus Oncolíticos/genética , Adenoviridae/genética , Antineoplásicos/toxicidad , Autofagia/efectos de los fármacos , Beclina-1/genética , Beclina-1/toxicidad , Línea Celular Tumoral , Doxorrubicina/toxicidad , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos
10.
J Hematol Oncol ; 10(1): 165, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-29058602

RESUMEN

T cells recognize antigen fragments from proteolytic products that are presented to them in the form of peptides on major histocompatibility complex (MHC) molecules, which is crucial for the T cell to identify infected or transformed cells. Autophagy, a process that delivers cytoplasmic constituents for lysosomal degradation, has been observed to provide a substantial source of intra- and extracellular antigens for MHC presentation to T cells, which will impact the tumor-specific immune response. Meanwhile, extracellular components are transported to cytoplasm for the degradation/secretion process by the endo-/exosomal pathway and are thus involved in multiple physiological and pathological processes, including immune responses. Autophagy and endo-/exosomal pathways are intertwined in a highly intricate manner and both are closely involved in antigen processing for MHC presentation; thus, we propose that they may coordinate in antigen processing and presentation in anticancer T cell immune responses. In this article, we discuss the molecular and functional crosstalk between autophagy and endo-/exosomal pathways and their contributions to antigen processing for MHC presentation in anticancer T cell immune responses.


Asunto(s)
Presentación de Antígeno/inmunología , Autofagia/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos
11.
Oncotarget ; 8(7): 12374-12388, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-27902471

RESUMEN

Autophagy is a tightly regulated catabolic process that leads to the degradation of cytoplasmatic components such as aggregated/misfolded proteins and organelles through the lysosomal machinery. Recent studies suggest that autophagy plays such a role in the context of the anti-tumor immune response, make it an attractive target for cancer immunotherapy. Defective autophagy in hematopoietic stem cells may contribute to the development of hematologic malignancies, including leukemia, myelodysplastic syndrome, and lymphoproliferative disorder. In blood cancer cells, autophagy can either result in chemoresistance or induce autophagic cell death that may act as immunogenic. Based on the successful experimental findings in vitro and in vivo, clinical trials of autophagy inhibitor such as hydroxychloroquine in combination with chemotherapy in patients with blood cancers are currently underway. However, autophagy inactivation might impair autophagy-triggered anticancer immunity, whereas induction of autophagy might become an effective immunotherapy. These aspects are discussed in this review together with a brief introduction to the autophagic molecular machinery and its roles in hematologic malignancies.


Asunto(s)
Inmunidad Adaptativa/inmunología , Autofagia/inmunología , Neoplasias Hematológicas/inmunología , Células Madre Neoplásicas/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Autofagia/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Inmunoterapia/métodos , Modelos Inmunológicos , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA