Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Am J Cancer Res ; 14(3): 1121-1138, 2024.
Article in English | MEDLINE | ID: mdl-38590396

ABSTRACT

Autophagy, a highly regulated lysosome-dependent catabolic pathway, has garnered increasing attention because of its role in leukemia resistance. Among the S100 family of small calcium-binding proteins, S100P is differentially expressed in various tumor cell lines, thereby influencing tumor occurrence, invasion, metastasis, and drug resistance. However, the relationship between S100P and autophagy in determining chemosensitivity in leukemia cells remains unexplored. Our investigation revealed a negative correlation between S100P expression and the clinical status in childhood leukemia, with its presence observed in HL-60 and Jurkat cell lines. Suppression of S100P expression resulted in increased cell proliferation and decreased chemosensitivity in leukemia cells, whereas enhancement of S100P expression inhibited cell proliferation and increased chemosensitivity. Additionally, S100P knockdown drastically promoted autophagy, which was subsequently suppressed by S100P upregulation. Moreover, the p53/AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway was found to be functionally associated with S100P-mediated autophagy. Knockdown of S100P expression led to a decrease in p53 and p-mTOR levels and an increase in p-AMPK expression, ultimately promoting autophagy. This effect was reversed by administration of Tenovin-6 (a p53 activator) and Compound C (an AMPK inhibitor). The findings of our in vivo experiments provide additional evidence supporting the aforementioned data. Specifically, S100P inhibition significantly enhanced the growth of HL-60 tumor xenografts and increased the expression of microtubule-associated protein 1 light chain 3 and p-AMPK in nude mice. Consequently, it can be concluded that S100P plays a regulatory role in the chemosensitivity of leukemia cells by modulating the p53/AMPK/mTOR pathway, which controls autophagy in leukemia cells.

2.
Heliyon ; 10(5): e27305, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38495131

ABSTRACT

Background: Limited research has been conducted on the impact of rituximab on immune function and the incidence of side effects in children undergoing combination chemotherapy for aggressive mature B-cell lymphoma/leukemia. Methods: Clinical data from 85 patients with primary pediatric aggressive mature B-cell lymphoma/leukemia, treated according to the Chinese Children's Cancer Group (CCCG)-mature B-cell non-Hodgkin lymphoma (BNHL)-2015 protocol from June 1, 2015, to December 1, 2022, were collected from three tertiary medical centers in China. Patients with pre-existing malignancies or primary immune deficiencies (PIDs) were excluded. Results: Between June 1, 2015, and December 1, 2022, 85 patients (65 [76.5%] boys and 20[23.5%] girls; mean age, 6.95 years) were enrolled, and immune data at baseline during follow-up were analyzed. At the end of chemotherapy, a higher proportion of patients in the R4 group exhibited a decrease in peripheral blood CD3- CD19+ B cells (20[100%] of 20 vs 13[47.8%] of 18, p = 0.04), CD3+ T cells (21[91.3%] of 23 vs 14[60.9%] of 23, p = 0.016), and serum IgM (14[60.9%] of 23 vs 4[17.4%] of 23, p = 0.003) compared to the R3 group. However, these differences were no longer statistically significant six months after chemotherapy administration. The combination of rituximab with AA was associated with a higher incidence of significant thrombocytopenia (49[81.7%] of 60 vs 29[52.7%] of 55, p = 0.001) and infection (35[58.3%] of 60 vs 17[30.9%] of 55, p = 0.003) compared to AA alone. Furthermore, the combination of rituximab with BB was linked to a higher incidence of significant thrombocytopenia (32[52.5%] of 61 vs 31[31.0%] of 100, p = 0.007) compared to BB alone. Conclusions: While the effects of rituximab in combination with intense chemotherapy for childhood aggressive mature B-cell lymphoma/leukemia on children's immune function generally recovers within six months it may still prolong the recovery from immunoglobulinemia, posing a risk of secondary infections. Further studies are required to identify children with potential primary immunodeficiencies.

3.
J Transl Med ; 21(1): 606, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679782

ABSTRACT

BACKGROUND: The identifying of B-cell lymphoma 2 (Bcl-2) as a therapeutic target has led to a paradigm shift in acute myeloid leukemia (AML) treatment. Pyroptosis is a novel antitumor therapeutic mechanism due to its cytotoxic and immunogenic effects. The combination of venetoclax and hypomethylating agents (HMAs) has been shown to lead to durable responses and significantly improve prognosis in patients with AML. However, our understanding of the mechanisms underlying this combinatorial activity is evolving. METHODS: We investigated whether the Bcl-2 inhibitor venetoclax induces AML cell pyroptosis and identified pyroptosis effector proteins. Via using western blotting, immunoprecipitation, RNA interference, CCK8 assays, and LDH assays, we explored the mechanism underlying the pyroptotic effect. The relationship between the expression of the pyroptosis effector protein GSDME and AML prognosis was investigated. The effect of GSDME demethylation combined with venetoclax treatment on pyroptosis was investigated and confirmed in mouse models and clinical samples. RESULTS: Venetoclax induces pyroptosis that is mediated by caspase-3-dependent GSDME cleavage. Mechanistically, venetoclax upregulates caspase-3 and GSDME cleavage by activating the intrinsic apoptotic pathway. GSDME is downregulated in AML by promoter methylation, and low GSDME expression is significantly associated with poor prognosis, based on public databases and patient sample analysis. In vivo and in vitro experiments showed that GSDME overexpression or HMAs-mediated restoration of GSDME expression significantly increased venetoclax-induced pyroptosis in AML. CONCLUSION: GSDME-mediated pyroptosis may be a novel aspect of the antileukemic effect of Bcl-2 inhibitors. This finding offers new insights into potential biomarkers and therapeutic strategies, identifying an important mechanism explaining the clinical activity of venetoclax and HMAs in AML.


Subject(s)
Biological Assay , Pyroptosis , Animals , Mice , Caspase 3 , Proto-Oncogene Proteins c-bcl-2
4.
J Cancer Res Clin Oncol ; 149(13): 11233-11245, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37358666

ABSTRACT

PURPOSE: Circular RNA (circRNA) is a type of novel non-coding RNA with close involvement in the tumorigenesis and treatment response of leukemias. This study aimed to screen and validate candidate circRNAs that estimate disease risk and response to induction therapy of pediatric acute myeloid leukemia (AML). METHODS: Bone marrow samples were obtained from 4 complete response (CR) pediatric AML patients, 4 non-CR pediatric AML patients, and 4 controls to screen differentially expressed circRNAs (DECs) through microarray analyses. Ten candidate circRNAs were selected and validated in 40 pediatric AML patients and 10 controls through reverse transcription-quantitative polymerase chain reaction. RESULTS: Microarray assay discovered 378 upregulated DECs and 688 downregulated DECs in pediatric AML patients vs. controls; 832 upregulated DECs and 950 downregulated DECs in CR AML patients vs. non-CR AML patients. Then cross-analysis identified 441 DECs that both related to pediatric AML risk and CR achievement. Further validation of ten candidate circRNAs in larger sample-sized populations showed that circ_0032891, circ_0076995, circ_0014352, circ_0047663, circ_0007444, circ_0001684, circ_0000544, and circ_0005354 were related to pediatric AML risk; circ_0032891, circ_0076995, circ_0014352, circ_0047663, circ_0007444, circ_0001684, and circ_0000544 were related to CR achievement in pediatric AML patients. Regarding the correlation of candidate circRNAs with survival profile, only circ_0032891, circ_0076995, and circ_0000544 forecasted event-free survival; circ_0076995 and circ_0001684 estimated overall survival in pediatric AML patients. CONCLUSION: CircRNA profile is intensively implicated in the disease risk and treatment response of pediatric AML, especially that circ_0032891, circ_0000544, circ_0076995, and circ_0001684 are related to pediatric AML risk, CR achievement, and survival.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Child , Humans , RNA, Circular/genetics , Microarray Analysis , Remission Induction , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics
5.
Am J Cancer Res ; 13(2): 436-451, 2023.
Article in English | MEDLINE | ID: mdl-36895972

ABSTRACT

Pyroptosis, a newly discovered mode of programmed cell death (PCD), is important in the regulation of cancer development. High mobility group box 1 (HMGB1) is a non-histone nuclear protein that is closely related to tumor development and chemotherapy resistance. However, whether endogenous HMGB1 regulates pyroptosis in neuroblastoma remains unknown. Here, we showed that HMGB1 showed ubiquitous higher expression in SH-SY5Y cells and clinical tumors, and was positively correlated with the risk factors of patients with neuroblastoma. Knockdown of GSDME or pharmacological inhibition of caspase-3 blocked pyroptosis and cytosolic translocation of HMGB1. Moreover, knockdown of HMGB1 inhibited cisplatin (DDP) or etoposide (VP16)-induced pyroptosis by decreasing GSDME-NT and cleaved caspase-3 expression, resulting in cell blebbing and LDH release. Knockdown of HMGB1 expression increased the sensitivity of SH-SY5Y cells to chemotherapy and switched pyroptosis to apoptosis. Furthermore, the ROS/ERK1/2/caspase-3/GSDME pathway was found to be functionally connected with DDP or VP16-induced pyroptosis. Hydrogen peroxide (H2O2, a ROS agonist) and EGF (an ERK agonist) promoted the cleavage of GSDME and caspase-3 in DDP or VP16 treatment cells, both of which were inhibited by HMGB1 knockdown. Importantly, these data were further supported by the in vivo experiment. Our study suggests that HMGB1 is a novel regulator of pyroptosis via the ROS/ERK1/2/caspase-3/GSDME pathway and a potential drug target for therapeutic interventions in neuroblastoma.

6.
Front Cell Infect Microbiol ; 12: 1002140, 2022.
Article in English | MEDLINE | ID: mdl-36339330

ABSTRACT

Mendelian susceptibility to mycobacterial diseases (MSMD) is a rare congenital immune deficiency characterized by susceptibility to weakly virulent mycobacteria. Loss-of-function (LOF) mutation of signal transducer and activator of transcription 1 (STAT1) is one of the common genetic causes of MSMD. In this study, we identified a patient who presented with multiple lymph node enlargements and multiple osteolytic disruptions. Mycobacterium gordonae infection was confirmed by metagenomic next-generation sequencing. Whole-exome sequencing identified a novel paternal heterozygous mutation in exon 22 of STAT1 (NM_007315.4, c.1892T>C, p.Val631Ala). This variant was confirmed pathogenic by multiple software predictions. Based on functional assays, STAT1 expression in STAT1V631A cells was not different from STAT1WT cells. But STAT1V631A mutation caused much lower activation of STAT1 when stimulated by interferon-γ (IFN-γ). Fluorescence localization analysis revealed that both STAT1V631A and STAT1WT proteins were located in the cytoplasm, and only a few STAT1V631A proteins were translocated to the nucleus in response to IFN-γ. These results suggest that STAT1V631A leads to LOF in IFN-γ-mediated mycobacterial immunity, resulting in MSMD. Treatment with antibiotics has achieved ideal disease control for this patient, and no adverse events occurred during follow-up. The STAT1 LOF deficiency is a genetic cause of MSMD, which should be considered in patients with mycobacterial disease, especially those with bone involvement.


Subject(s)
Mycobacterium Infections , Mycobacterium , Humans , Genetic Predisposition to Disease , Mycobacterium Infections/genetics , Mutation , Interferon-gamma/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 24(9): 1036-1041, 2022.
Article in Chinese | MEDLINE | ID: mdl-36111723

ABSTRACT

OBJECTIVES: To study the clinical features of children with rhabdomyosarcoma (RMS) and the influencing factors for prognosis. METHODS: A retrospective analysis was performed on the clinical and follow-up data of 20 children with RMS who were admitted to the Department of Pediatric Hematology, Xiangya Hospital of Central South University, from June 2014 to September 2020. RESULTS: The most common clinical symptoms of the 20 children with RMS at the first visit were painless mass (13/20, 65%), exophthalmos (4/20, 20%), and abdominal pain (3/20, 15%). According to the staging criteria of Intergroup Rhabdomyosarcoma Study Group (IRSG), there was 1 child (5%) with stage I RMS, 4 (20%) with stage II RMS, 9 (45%) with stage III RMS, and 6 (30%) with stage IV RMS. The median follow-up time was 19 months for the 20 children (range: 3-93 months), with a 2-year overall survival (OS) rate of 79.5% (95%CI: 20.1-24.3) and a 2-year event-free survival (EFS) rate of 72.0% (95%CI: 19.5-23.9). Pleomorphic RMS was associated with the reduced 2-year OS rate (P<0.05), and distant metastasis, IRSG stage IV RMS, and high-risk RMS were associated with the reduced 2-year EFS rate (P<0.05). CONCLUSIONS: RMS has no specific clinical symptoms at the first visit, with painless mass as the most common symptom. Distant metastasis, IRSG stage, and risk degree may be associated with the prognosis of children with RMS.


Subject(s)
Rhabdomyosarcoma, Embryonal , Rhabdomyosarcoma , Child , Humans , Prognosis , Retrospective Studies , Rhabdomyosarcoma/diagnosis , Rhabdomyosarcoma/therapy , Survival Rate
8.
Discov Oncol ; 13(1): 89, 2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36114893

ABSTRACT

PURPOSE: The angiogenesis is among the primary factors that affect tumor recurrence and distant organ metastasis in colorectal cancer (CRC). N6-methyladenosine (m6A) modification is one of the most common chemical modifications in eukaryotic mRNA, especially at the post-transcriptional level. Methyltransferase-like 3 (METTL3) promoting angiogenesis in a variety of tumors has been reported. However, the mechanism of how METTL3 dual-regulates the stability of long non-coding RNAs (lncRNAs) and vascular-related factor RNAs to affect angiogenesis in CRC is unclear. METHODS: 64 paired CRC and adjacent normal tissues were collected. In vitro, quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), actinomycin assay, methylated RNA immunoprecipitation (MeRIP) experiment,3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and colony formation assay were performed. The functions were also studied in zebrafish model animals in vivo. RESULTS: We found that the vascular endothelial growth factor A(VEGFA), METTL3 and LINC00662 RNAs were highly expressed in CRC, and that METTL3 was significantly positively correlated with LINC00662 and VEGFA. The protein expression levels of CD31, CD34, VEGFA, m6A and METTL3 were all significantly increased in the CRC tissues. The angiogenesis experiments both in vivo and in vitro found that METTL3 and LINC00662 promoted angiogenesis in CRC. The actinomycin assay indicated that METTL3 maintained the stability of LINC00662 and VEGFA RNAs. In addition, the MeRIP experiment confirmed that the LINC00662 and VEGFA RNAs had METTL3-enriched sites. CONCLUSION: These findings suggest that METTL3 and LINC00662 may both serve as diagnostic and prognostic predictive biomarkers for CRC and potential targets for anti-vascular therapy.

9.
Clin Chim Acta ; 521: 212-214, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34270954

ABSTRACT

INTRODUCTION: Anti-IgLON5 disease is a rare autoimmune disease of the central nervous system. Different from the previous autoimmune encephalitis, the disease is a chronic progressive disease characterized by abnormal sleep, sleep apnea and motor disorders, which is prone to misdiagnosis and missed diagnosis. METHODS: We report a unique case of anti-IgLON5 disease in a pediatric patient with Langerhans cell histiocytosis (LCH). He gradually developed increased muscle tone and nystagmus during chemotherapy and showed signs of meningeal enhancement on cranial imaging. Due to insufficient evidence of LCH invasion of the central nervous system, the presence of autoimmune encephalitis-related antibodies was investigated by using cell-based assay (CBA) experiment in indirect immuno-fluorescence assay (IFA). RESULTS: Clinical manifestations of sleep disorders and motor disorders, plus the presence of IgLON5 IgG antibodies (1:30) in the serum leading to a confirmed diagnosis of anti-IgLON5 disease. CONCLUSION: Anti-IgLON5 disease is rare and almost no cases of children have been reported. In view of the difficult to recognize symptoms in pediatric patients, especially those with other comorbidities. Clinicians should raise their awareness of this disease and pay attention to the detection of autoimmune antibodies.


Subject(s)
Encephalitis , Hashimoto Disease , Histiocytosis, Langerhans-Cell , Cell Adhesion Molecules, Neuronal , Child , Histiocytosis, Langerhans-Cell/complications , Histiocytosis, Langerhans-Cell/diagnosis , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...