Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 111(11): 4051-4060, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32876369

RESUMEN

Ibrutinib might improve the efficacy of anti-CD19 chimeric antigen receptor (CD19 CAR) T-cell therapy in chronic lymphocytic leukemia (CLL). We studied the possibility and mechanism of the synergistic effect of ibrutinib and CAR-T cells in other types of lymphoma. In this study, we selected the CD19 CAR-T cells of a patient with lymphoma who failed in his CD19 CAR-T-cell therapy and a dose of 8 mg/kg/d ibrutinib. Subcutaneous and tail vein tumorigenic mice were established with Raji cells. The differences in the synergistic effect between these 2 models were compared by bioluminescence imaging (BLI) monitoring and flow cytometry (FCM). The expression of the STAT-3 signaling pathway was assessed by western blot analysis. There was no synergistic effect of ibrutinib and CD19 CAR-T cells in vitro. Programmed cell death-ligand 1 (PD-L1) was expressed in 0.23 ± 0.06% of Raji cells. In the subcutaneous tumorigenic model, the luciferase signal was reduced significantly in the group receiving ibrutinib combined with CD19 CAR-T cells. Moreover, the proportion of CD19 CAR-T cells was higher in the polytherapy group than in the CAR-T-cell monotherapy group. However, we did not get an analogous synergistic effect in the tail vein tumorigenic model. STAT-3 signaling pathway expression in the residual tumor cells did not differ between those with and those without ibrutinib, suggesting that the IL-10/STAT-3/PD-L1 pathway was not involved in the synergistic effect. Therefore, some other mechanism might be a target for ibrutinib. Our results provide evidence for the use of ibrutinib in polytherapy for other types of B-cell lymphoma.


Asunto(s)
Adenina/análogos & derivados , Antígenos CD19/inmunología , Inmunoterapia Adoptiva , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adenina/farmacología , Adulto , Anciano , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/terapia , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Transcripción STAT3/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cancer Cell Int ; 20: 375, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32782434

RESUMEN

BACKGROUND: The aim of this study is to determine whether Hypoxanthine Guanine Phosphoribosyltransferase (HPRT) could be used as a biomarker for the diagnosis and treatment of B cell malignancies. With 4.3% of all new cancers diagnosed as Non-Hodgkin lymphoma, finding new biomarkers for the treatment of B cell cancers is an ongoing pursuit. HPRT is a nucleotide salvage pathway enzyme responsible for the synthesis of guanine and inosine throughout the cell cycle. METHODS: Raji cells were used for this analysis due to their high HPRT internal expression. Internal expression was evaluated utilizing western blotting and RNA sequencing. Surface localization was analyzed using flow cytometry, confocal microscopy, and membrane biotinylation. To determine the source of HPRT surface expression, a CRISPR knockdown of HPRT was generated and confirmed using western blotting. To determine clinical significance, patient blood samples were collected and analyzed for HPRT surface localization. RESULTS: We found surface localization of HPRT on both Raji cancer cells and in 77% of the malignant ALL samples analyzed and observed no significant expression in healthy cells. Surface expression was confirmed in Raji cells with confocal microscopy, where a direct overlap between HPRT specific antibodies and a membrane-specific dye was observed. HPRT was also detected in biotinylated membranes of Raji cells. Upon HPRT knockdown in Raji cells, we found a significant reduction in surface expression, which shows that the HPRT found on the surface originates from the cells themselves. Finally, we found that cells that had elevated levels of HPRT had a direct correlation to XRCC2, BRCA1, PIK3CA, MSH2, MSH6, WDYHV1, AK7, and BLMH expression and an inverse correlation to PRKD2, PTGS2, TCF7L2, CDH1, IL6R, MC1R, AMPD1, TLR6, and BAK1 expression. Of the 17 genes with significant correlation, 9 are involved in cellular proliferation and DNA synthesis, regulation, and repair. CONCLUSIONS: As a surface biomarker that is found on malignant cells and not on healthy cells, HPRT could be used as a surface antigen for targeted immunotherapy. In addition, the gene correlations show that HPRT may have an additional role in regulation of cancer proliferation that has not been previously discovered.

3.
Arch Toxicol ; 94(9): 2997-3012, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32592077

RESUMEN

The universal presence of micro-nanoplastics (MNPLs) and its relative unknown effects on human health is a concern demanding reliable data to evaluate their safety. As ingestion is one of the main exposure routes for humans, we have assessed their hazard using two in vitro models that simulate the human intestinal barrier and its associated lymphoid system. Two different coculture models (differentiated Caco-2/HT29 intestinal cells and Caco-2/HT29 + Raji-B cells) were exposed to polystyrene nanoparticles (PSNPs) for 24 h. Endpoints such as viability, membrane integrity, NPS localization and translocation, ROS induction, and genotoxic damage were evaluated to have a comprehensive view of their potentially harmful effects. No significant cytotoxic effects were observed in any of the analyzed systems. In addition, no adverse effects were detected in the integrity or in the permeability of the barrier model. Nevertheless, confocal microscopy analysis showed that MNPLs were highly uptaken by both of the barrier model systems, and that translocation across the membrane occurred. Thus, MNPLs were detected into Raji-B cells, placed in the basolateral compartment of the insert. The internalization followed a dose-dependent pattern, as assessed by flow cytometry. Nonetheless, no genotoxic or oxidative DNA damage induction was detected in either case. Finally, no variations in the transcription of oxidative and stress genes could be detected in any of the in vitro barrier models. Our results show that MNPLs can enter and cross the epithelial barrier of the digestive system, as demonstrated when Raji-B cells were included in the model, but without exerting apparent hazardous effects.


Asunto(s)
Intestinos , Microplásticos/toxicidad , Poliestirenos/toxicidad , Células CACO-2 , Diferenciación Celular , Daño del ADN , Células HT29 , Humanos , Microscopía Confocal , Nanopartículas , Permeabilidad
4.
Saudi Pharm J ; 28(12): 1686-1703, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33424261

RESUMEN

The genus Millettia belongs to Fabaceae includes 200 species which are distributed in tropical and subtropical regions of the world. Plants belong to this genus are used as folkloric medicine, for the treatment of different ailments like in wound healing, boil, sores, skin diseases, snake bite, muscle aches, pains, rheumatic arthritis, and gynaecological diseases. The aim of the review is to provide updated, comprehensive and categorized information on the aspects of ethnobotanical, phytochemical, pharmacological uses and toxicity of genus Millettia in order to identify their therapeutic potential and generate space for future research opportunities. The present study comprises of isolated flavonoids, phenolic compounds, phytosterols, saponins, alkaloids, polysaccharides, terpenoids and resins and pharmacological activities of various Millettia species. The relevant data were searched by using the keyword "Millettia" in different scientific databases like, "Google Scholar"; "NISCAIR repository"; "Pub Med"; "Science Direct"; "Scopus" and the taxonomy is validated by "The Plant List". This review discusses the existing information of the traditional evaluation as well as phytochemical and pharmacological evaluation of the extract and active constituents of the genus "Millettia". This review confirms that several Millettia species have emerged as a high-quality medicine in a traditional system for arthritis, wound healing, inflammation, skin diseases. Numerous conventional uses of Millettia species have been validated by modern pharmacology research. Intensive investigations of the genus Millettia relating to phytochemistry and pharmacology, especially their mechanism of action, safety, and efficacy could be the future research interests by the researcher in the area of phytomedicine.

5.
Bull Exp Biol Med ; 166(6): 785-787, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31028581

RESUMEN

We studied the effect of low and high-dose rate photon radiation on activation of cell death by apoptosis and necrosis in malignant cell lines of lymphocytic origin Raji and Jurkat (human B and T-cell lymphomas) and normal human lymphocytes from healthy volunteers. It was shown that photon radiation with ultra-high dose rate induced significantly higher levels of "early" apoptosis and lower levels of necrosis compared to γ-radiation with dose rate used for radiation therapy.


Asunto(s)
Apoptosis/efectos de la radiación , Rayos gamma , Linfocitos/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Células Jurkat , Linfocitos/patología , Necrosis/patología , Cultivo Primario de Células
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(3): 305-310, 2019 May.
Artículo en Zh | MEDLINE | ID: mdl-31631594

RESUMEN

OBJECTIVE: To determine the effects of three histone methylase inhibitors UNC1999, DZNep and GSK343 on the survival, apoptosis and cell cycle of non-hodgkin's lymphoma Raji cells. METHODS: PCR amplified 16 and 18 exons of enhancer of zeste homolog 2 ( EZH2) gene were detected. The expression of EZH2 in normal adult lymphocytes and Raji cells was detected by Western blot. The Raji cells were treated by UNC1999, DZNep and GSK343, followed by CCK-8 assays analyzing cell survival, flow cytometry detecting cell apoptosis and cell cycle, and Western blot detecting the expressions of EZH2 and H3K27 me3. RESULTS: The Sanger sequencing results showed that the Raji cells did not carry Y641 and A677 mutation sites of EZH2. The Western blot results showed high expressions of EZH2 in the Raji cells. The results of CCK-8 showed that UNC1999, DZNep and GSK343 inhibited cell survival, and the weakest effect was from DZNep. The flow cytometric assay showed that UNC1999, DZNep and GSK343 promoted apoptosis of the Raji cells, and the effect of UNC1999 was stronger than that of GSK343 and DZNep. The cell cycle was arrested at phase G 1/G 0 after treatment of the Raji cells with the three inhibitors, with UNC1999 triggering the most significant changes. The Western blot showed that UNC1999 and GSK343 inhibited the histone methylase activity of EZH2 and significantly reduced the expression of H3K27 me3. CONCLUSION: EZH2 inhibitors can inhibit cell survival, promote cell apoptosis and arrest cell cycle at phase G 1/G 0 of Raji cells through reducing the expression of H3K27me3. UNC1999 has a stronger effect than GSK343 and DZNep.


Asunto(s)
Apoptosis , Puntos de Control del Ciclo Celular , Histona Metiltransferasas/antagonistas & inhibidores , Complejo Represivo Polycomb 2 , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Indazoles/farmacología , Linfocitos , Piridonas/farmacología
7.
Cell Biol Int ; 42(11): 1503-1510, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30039885

RESUMEN

In human lung cancer, Tripartite motif 65 (TRIM65) is documented as an important regulator in carcinogenesis. Knockdown of TRIM65 prevents the tumorigenesis of lung cancer cells, while TRIM65 overexpression presents the opposite effect. However, the roles of TRIM65 in human lymphocyte malignancies have reported little. Herein, we found that Jurkat (T-lymphocyte) and Raji (B-lymphocyte) expressed TRIM65. We aimed to investigate whether TRIM65 was a potential oncogenic protein that regulated the tumorigenesis of Jurkat and Raji cells. In our present study, cells were transfected with siRNA-TRIM65 or TRIM65 overexpression vector, Cell counting kit-8 (CCK-8), Flow cytometry and Annexin V-FITC/propidium iodide (PI) staining was carried out to detect cell viability, cell cycle profile and cell apoptosis, respectively. Extracellular signal-regulated kinases 1/2 (ERK1/2) pathway-associated proteins, such as Bcl2, cleaved-caspase 3, vascular endothelial growth factor (VEGF), and phosphorylated ERK1/2 (p-ERK1/2) were assessed. Our data indicated that knockdown of TRIM65 prevented the tumorigenesis of Jurkat and Raji cells. TRIM65 silencing inhibited cell proliferation, promoted cell apoptosis and arrested cell cycle, highly like through blocking ERK1/2 pathway. However, TRIM65 overexpression enhanced cell viability, increased the protein levels of Bcl2, VEGF, p-ERK1/2 while decreased cleaved-caspase 3 expression, suggesting the promoted effect of TRIM65 overexpression in the tumorigenesis of those two lymphoma cells. To validate the involvement of ERK1/2 pathway, ERK1/2 inhibitor AZD8330 (1 µmol/L) was introduced. We found that AZD8330 significantly prevented TRIM65 overexpression-induced tumorigenesis. We concluded that TRIM65 served as a potential oncogenic protein on Jurkat and Raji cells, and ERK1/2 pathway was the underlying mechanism. Approaches targeting TRIM65 provided a novel strategy for the treatment of lymphoma.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Linfoma/metabolismo , Linfoma/patología , Terapia Molecular Dirigida , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Apoptosis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dihidropiridinas/farmacología , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Células Jurkat , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
8.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28630073

RESUMEN

Salmonella enterica serovar Typhi causes the systemic disease typhoid fever. After ingestion, it adheres to and invades the host epithelium while evading the host innate immune response, causing little if any inflammation. Conversely, Salmonella enterica serovar Typhimurium causes gastroenteritis in humans and thrives in the inflamed gut. Upon entering the host, S Typhimurium preferentially colonizes Peyer's patches, a lymphoid organ in which microfold cells (M cells) overlay an arrangement of B cells, T cells, and antigen-presenting cells. Both serovars can adhere to and invade M cells and enterocytes, and it has been assumed that S Typhi also preferentially targets M cells. In this study, we present data supporting the alternative hypothesis that S Typhi preferentially targets enterocytes. Using a tissue culture M cell model, we examined S Typhi strains with a deletion in the stg fimbriae. The stg deletion resulted in increased adherence to M cells and, as expected, decreased adherence to Caco-2 cells. Adherence to M cells could be further enhanced by introduction of the long polar fimbriae (Lpf), which facilitate adherence of S Typhimurium to M cells. Deletion of stg and/or introduction of lpf enhanced M cell invasion as well, leading to significant increases in secretion of interleukin 8. These results suggest that S Typhi may preferentially target enterocytes in vivo.


Asunto(s)
Adhesión Bacteriana , Enterocitos/microbiología , Fimbrias Bacterianas/metabolismo , Salmonella typhi/fisiología , Células CACO-2 , Humanos
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 239-252, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27641442

RESUMEN

B-cells of the high-grade non-Hodgkin lymphoma Burkitt's lymphoma (BL) overexpress survival oncoproteins, including the proviral integration site for Moloney murine leukaemia virus kinase (Pim)-1, and become apoptosis resistant. Activated death receptor CD95 after ligation with anti-CD95 monoclonal antibody (mAb) resulted in the regression of BL via induction of apoptosis, suggesting a decrease of survival protein expression. Here, CD95-mediated apoptotic pathways in BL B-cell lines (Raji and Daudi) following treatment with anti-CD95 mAb was investigated with the cause-and-effects on pim-1 gene expression, in comparison with leukemic cell line (K562) used as CD95-negative cells. Immunohistochemical staining for CD95 and Pim-1 was performed, and the effects of anti-CD95 mAb on apoptotic signalling using western blotting, on caspase activity and cell survival of BL B-cell and leukemic cell lines were determined. We showed that Raji cells expressed more CD95 receptors than Daudi cells. Half of each population underwent apoptosis accompanied by decreased cell viability after anti-CD95 mAb treatment. Distinct extrinsic and intrinsic CD95-mediated apoptotic pathways in Raji and Daudi cells were revealed by high caspase activity and mitochondrial outer membrane permeabilization, respectively. We observed decreased Pim-1 transcript and protein expression levels with increased heat-shock protein (Hsp)70 and decreased Hsp90 expression in anti-CD95 mAb-treated cells. Throughout the study, K562 cells did not undergo apoptosis upon anti-CD95 mAb treatment. Pim-1 knockdown following to stable transfection with plasmid vectors induced apoptosis and decreased viability of BL and K562 cells. Therefore, CD95-mediated apoptosis induces Pim-1 down-regulation in BL B-cells, but Pim-1 down-regulation cannot fully eradicate BL and leukaemia.


Asunto(s)
Apoptosis , Linfoma de Burkitt/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-pim-1/genética , Receptor fas/metabolismo , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
10.
Regul Toxicol Pharmacol ; 85: 79-85, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28185844

RESUMEN

Glyphosate is a highly used active compound in agriculturally based pesticides. The literature regarding the toxicity of glyphosate to human cells has been highly inconsistent. We studied the resulting DNA damage and cytotoxicity of various glyphosate concentrations on human cells to evaluate DNA damaging potential. Utilizing human Raji cells, DNA damage was quantified using the comet assay, while cytotoxicity was further analyzed using MTT viability assays. Several glyphosate concentrations were assessed, ranging from 15 mM to 0.1 µM. We found that glyphosate treatment is lethal to Raji cells at concentrations above 10 mM, yet has no cytotoxic effects at concentrations at or below 100 µM. Treatment concentrations of 1 mM and 5 mM induce statistically significant DNA damage to Raji cells following 30-60 min of treatment, however, cells show a slow recovery from initial damage and cell viability is unaffected after 2 h. At these same concentrations, cells treated with additional compound did not recover and maintained high levels of DNA damage. While the cytotoxicity of glyphosate appears to be minimal for physiologically relevant concentrations, the compound has a definitive cytotoxic nature in human cells at high concentrations. Our data also suggests a mammalian metabolic pathway for the degradation of glyphosate may be present.


Asunto(s)
Glicina/análogos & derivados , Herbicidas/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN , Relación Dosis-Respuesta a Droga , Glicina/toxicidad , Humanos , Glifosato
11.
Arch Pharm (Weinheim) ; 350(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28960496

RESUMEN

Cladribine (2-CdA) is used as an anti-cancer drug but is currently studied as a potential treatment for use in relapsing-remitting multiple sclerosis (MS). In this study, we computer designed, synthesized, and characterized two novel derivatives of 2-CdA, K1-5d and K2-4c, and investigated their underlying mechanism of beneficial effect using the CCRF-CEM and RAJI cell lines. For this purpose, we first determined their effect on MS and DNA damage and repair-related gene expression profiles using custom arrays along with 2-CdA treatment at non-toxic doses. Then, we determined whether cells underwent apoptosis after treatment with 2-CdA, K1-5d, and K2-4c in CCRF-CEM and RAJI cells, using the DNA fragmentation assay. It was found that both derivatives modulated the expression of the pathway-related genes that are important in inflammatory signaling, apoptosis, ATM/ATR, double-strand break repair, and the cell cycle. Furthermore, 2-CdA, K1-5d, and K2-4c significantly activated apoptosis in both cell lines. In summary, our data demonstrate that although both derivatives act as anti-inflammatory and apoptotic agents, inducing the accumulation of DNA strand breaks and activating the ultimate tumor suppressor p53 in T and B lymphocytes, the K1-5d derivative has shown more promising activities for further studies.


Asunto(s)
Cladribina/farmacología , Daño del ADN/efectos de los fármacos , Inmunosupresores/farmacología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Cladribina/síntesis química , Cladribina/química , Simulación por Computador , Roturas del ADN/efectos de los fármacos , Humanos , Inmunosupresores/síntesis química , Inmunosupresores/química , Simulación del Acoplamiento Molecular , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
12.
Oncol Rep ; 51(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38275109

RESUMEN

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that a pair of the lanes featured in the western blot gel for Fig. 3B on p. 1705 were strikingly similar, suggesting that the same data may have been imported into the figure to represent data that were intended to show the results from differently performed experiments. In addition, the western data shown in Fig. 6 on p. 1706 for the GRP78 and the p­PERK proteins also appeared to be strikingly similar, such that the same data may have also been inserted into this figure to show the results from different experiments. Finally, certain of the data in this paper were shown to have appeared in a previous publication that featured some of the same authors, albeit in different form [Zhang L, Huang D, Wang Q, Shen D, Wang Y, Chen B, Zhang J and Gai L: MiR­132 inhibits expression of SIRT1 and induces pro­inflammatory processes of vascular endothelial inflammation through blockade of the SREBP­1c metabolic pathway. Cardiovasc Drugs Ther 8: 303­311, 2014]. Owing to the fact that some of the contentious data in the above article had already been published elsewhere prior to its submission to Oncology Reports, and because of an overall lack of confidence in the presented data, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 36: 1702­1708, 2016; DOI: 10.3892/or.2016.4975].

13.
Int J Mol Cell Med ; 12(1): 1-17, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942260

RESUMEN

Leukemia is a type of cancer that affects the blood and bone marrow. Acute lymphoid leukaemia, also known as ALL, is regarded as one of the deadliest forms of cancer. Due to the rapid increase in various cancer cases and the development of resistance in cancer cells, it is necessary to identify novel lead molecules with more potent anticancer properties. There is a growing interest in using herbal products/analogs as multi-component agents (as anticancer agents and immunomodulators) for cancer treatment. In the present investigation, an attempt has been made to explore the anticancer and immunomodulatory activity of P19, an analog of parthenin in ALL. P19 was reported to exhibit anticancer efficacy by triggering apoptotic signaling events in human leukaemia HL-60 cells by significant NO production. In contrast to this finding, ROS and NO were not required for P19-mediated apoptosis in Raji cells. The mechanism of action of P19 was observed to be cancer cell lineage dependent. P19 demonstrated very effective anticancer properties against ALL (IC50 3µM). Molecular investigations revealed that P19 induced mitochondrion mediated apoptosis by Bax localization to mitochondria and enhanced cytosolic calcium in the cytoplasm. Further activation of the caspase 3, caspase 8 and PARP cleavage suggested the involvement of the caspase-mediated apoptosis. Anti-proliferative activity revealed the telomerase inhibition and cell cycle arrest in G0/G1 phase after P19 treatment. Immunomodulatory effects of the P19 revealed the enhanced INFÉ£ and NO production in Jurkat and THP cells. Owing to its antiproliferative and immunomodulatory potential against leukemia cells P19 can further be explored as effective therapeutics against leukemia.

14.
Vet Immunol Immunopathol ; 264: 110658, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37748249

RESUMEN

The infectious bursal disease virus (IBDV) causes an acute and highly contagious immunosuppressive response in young chickens by targeting B lymphocytes in immune organs. Changes in regulatory T-cell ratio and apoptosis have been demonstrated during IBDV infection in these cells. The possible change in CD19 expression as the precursor of B cells after IBDV replication was detected in this study. Raji cells were infected with an IBDV isolate at MOIs of 1.0 and 3.0. The viral kinetics were determined using the characteristic virus-induced CPE, cell viability, and infectious titer. Induction of apoptosis and also changes in the CD19 expression within the virus infection were assessed by flow cytometry. The Raji cells were found to be susceptible to IBDV infection by producing marked CPEs dependent on MOI. The infectivity titers were determined in intra- and extracellular samples at the defined hours. The kinetics of early IBDV replication in Raji cells were nearly identical for both MOIs, but a significant difference in the infectivity titer was observed at 48 hpi. The quick apoptotic events were observed to be significantly higher in MOI 3.0, which was correlated with the lower virus titer. A significant CD19 expression change in the IBDV-infected Raji cells was revealed. The results suggested that Raji cells mimic the IBDV replication in lymphoid organs and the virus replication is related to CD19 expression frequencies in the lymphoid cells.


Asunto(s)
Infecciones por Birnaviridae , Virus de la Enfermedad Infecciosa de la Bolsa , Enfermedades de las Aves de Corral , Animales , Pollos , Linfocitos B , Linfocitos , Replicación Viral , Infecciones por Birnaviridae/veterinaria
15.
Chemosphere ; 325: 138360, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36905991

RESUMEN

The environmental presence of micro/nanoplastics (MNPLs) is an environmental and human health concern. Such MNPLs can result from the physicochemical/biological degradation of plastic goods (secondary MNPLs) or can result from industrial production at that size, for different commercial purposes (primary MNPLs). Independently of their origin, the toxicological profile of MNPLs can be modulated by their size, as well as by the ability of cells/organisms to internalize them. To get more information on these topics we have determined the ability of three different sizes of polystyrene MNPLs (50, 200, and 500 nm) to produce different biological effects in three different human hematopoietic cell lines (Raji-B, THP-1, and TK6). Results show that none of the three sizes was able to induce toxicity (growth ability) in any of the tested cell types. Although transmission electron microscopy and confocal images showed cell internalization in all the cases, their quantification by flow cytometry demonstrated an important uptake by Raji-B and THP-1 cells, in comparison with TK6 cells. For the first ones, the uptake was negatively associated with the size. Interestingly, when the loss of mitochondrial membrane potential was determined, dose-related effects were observed for Raji-B and THP-1 cells, but not for TK6 cells. These effects were observed for the three different sizes. Finally, when oxidative stress induction was evaluated, no clear effects were observed for the different tested combinations. Our conclusion is that size, biological endpoint, and cell type are aspects modulating the toxicological profile of MNPLs.


Asunto(s)
Nanopartículas , Poliestirenos , Humanos , Poliestirenos/toxicidad , Microplásticos/toxicidad , Plásticos/toxicidad , Línea Celular , Nanopartículas/toxicidad
16.
Bio Protoc ; 13(8): e4655, 2023 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-37113332

RESUMEN

Chimeric antigen receptor (CAR)-T therapy launched a new era for cancer treatments, displaying outstanding effectiveness in relapsed or refractory B-cell malignancies. Demonstrating the tumor-killing ability of CAR-Ts in mouse xenograft models serves as a golden criterium in preclinical research. Here, we describe a detailed method for evaluating CAR-T's function in immune-deficient mice bearing Raji B cell-induced tumors. It includes generating CD19 CAR-T cells from healthy donors, injecting tumor cells and CAR-T cells into mice, and monitoring tumor growth and CAR-T state. This protocol provides a practical guide to evaluate CAR-T's function in vivo within eight weeks. Graphical abstract.

17.
Cancers (Basel) ; 15(23)2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-38067224

RESUMEN

Very Small Embryonic-like Stem Cells (VSELSCs) and Very Small Cancer Stem Cells (VSCSCs) are fields of intensive research. Although the presence in vitro of VSELSC and VSCSC cellular stage analogs appear probable, it has yet to be published. Utilizing established human cell cultures with varying populations of primitive cells, stained with CD markers specific to primitive stages, in addition to a fluorescent DNA dye, and following histochemical processing, we have developed a cytological method for detecting Very Small Leukemic Stem-like Cells (VSLSLCs), Very Small Cancer Stem-like Cells (VSCSLCs), and VSELSCs. This detection provides an opportunity to advance research in these areas.

18.
Biomed Pharmacother ; 156: 113887, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36274467

RESUMEN

Lymphoma is a cancer of the lymphoid cells that originated in matured B or T cells. The bioactive natural compounds can efficiently treat this disease with lesser side effects. Thus, in this study, a natural stilbene B10 (3-methoxy 5-hydroxy stilbene) isolated from Cajanus cajan (Pigeon Pea) was screened for its anti-proliferative efficacy against 13 cancer cell lines. B10 showed a potential effect on the human lymphoma (Raji) cells. Cytotoxicity analysis of B10 has revealed IC50 concentrations in Raji cells at low doses (18 µM) than other cancer cell lines. The B10 could significantly cause dose and time-dependent inhibition in the proliferation of Raji cells triggering intrinsic apoptosis and S/G1 phase cellular arrest. There was an increased expression of phospho-γ-H2A.X and decreased expression of cyclin D1, causing DNA damage and cell cycle arrest, post- B10 treatments. The mitochondrial membrane potential (MMP) variations observed after B10 treatment led to changes in Bax/Bcl-2 ratio, cytochrome C release, and enhanced expression of cleaved caspase3, 9, PARP-1, and APAF-1. The B10 inhibited the proliferation of Raji cells by significantly downregulating the expression of KRAS, BTK, MDM2, P-JAK2, P-STAT3, PI3K, HDAC1/2, SIRT7, and EP300. The treatment upregulated the tumor suppressor genes PEBP1 and SAP18. Thus, the study could reveal the selective inhibitory effects of B10 on lymphoma, suggesting it as a probable innovative chemotherapeutic agent.


Asunto(s)
Estilbenos , Humanos , Estilbenos/farmacología , Proteínas Proto-Oncogénicas p21(ras) , Proliferación Celular , Línea Celular Tumoral , Apoptosis , Linfocitos , Proteínas de Unión a Fosfatidiletanolamina , Histona Desacetilasa 1 , Proteína p300 Asociada a E1A
19.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(6): 1746-1751, 2022 Dec.
Artículo en Zh | MEDLINE | ID: mdl-36476898

RESUMEN

OBJECTIVE: To investigate the effect of atovaquone on the cell cycle and apoptosis of non-Hodgkin's lymphoma Raji cells, and clarify the related mechanisms. METHODS: MTT assay and trypan blue dye exclusion method were used to evaluate the effect of atovaquone on the proliferation of Raji cells. After the cells were stained by PI staining, the cell cycle distribution was detected by flow cytometry. Cell apoptosis was analyzed by Annexin V/PI double binding assay. The intracellular alterations of reactive oxygen species were detected by 2', 7'-dichlorofluorescein diacetate (DCFH-DA). The protein expression of cell cycle and apoptosis related molecules were detected by Western blot. RESULTS: Various concentrations of atovaquone (5-40 µmol/L) inhibited the growth of Raji cells in a concentration-dependent manner (r=0.951). The proliferation of Raji cells was significantly inhibited after treated by atovaquone (20 and 30 µmol/L) for 24, 48 and 72 h, which showed statistically different with that in the control group (P<0.01, P<0.001, P<0.001). G1 phase arrest (P<0.01, P<0.001) and apoptosis (P<0.01) of Raji cells was induced by atovaquone (20 and 30 µmol/L) significantly for 24 h and 48 h, respectively. The expression of p-JAK2 and p-STAT3(Y705) protein were down-regulated significantly induced by atovaquone (P<0.001, P<0.05). Furthermore, atovaquone treatment could induce the decreasing of antiapoptotic protein Mcl-1, Bcl-2, and Bcl-xl expression level (P<0.05) and increasing of cleaved caspase-3 protein expression level. In addition, atovaquone could also induce the down-regulation of c-Myc (P<0.001, P<0.01) and cell cycle related molecules Cyclin D1, CDK4, and CDK6 (P<0.01, P<0.05) protein expression. CONCLUSION: Atovaquone effectively inhibits cell proliferation and induces cell cycle arrest and apoptosis by suppression of STAT3 signaling pathway in Raji cells. It can be a potential therapeutic agent against non-Hodgkin's lymphoma.


Asunto(s)
Apoptosis , Linfoma no Hodgkin , Humanos , Atovacuona/farmacología , Puntos de Control del Ciclo Celular
20.
Oncol Lett ; 21(1): 56, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33281967

RESUMEN

The incidence of non-Hodgkin's lymphoma (NHL) has been increasing annually and has become a serious threat to human health. However, the pathogenesis of NHL remains unclear. The present study aimed to investigate the effect of soluble CD40 ligand (sCD40L) on NHL cells and its underlying mechanism. Cell Counting kit-8 assay and flow cytometry apoptosis experiments were conducted to investigate the effects of sCD40L on cell proliferation and apoptosis. Western blotting was performed to detect the protein expression levels of BAX, Bcl-2, ERK, p-ERK, JNK, p-JNK, p38, p-p38 and c-JUN. The results of the present study demonstrated that exogenous sCD40L significantly inhibited the proliferation and promoted the apoptosis of Raji and CA46 cells. Additionally, exogenous sCD40L promoted the apoptosis of lymphoma cells by activating the JNK signaling pathway.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA