Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Leuk Res ; 138: 107453, 2024 03.
Article in English | MEDLINE | ID: mdl-38442594

ABSTRACT

Familial acute myeloid leukemia (AML) pedigrees with germline CCAAT/enhancer-binding protein-α (CEBPA) mutation have been rarely reported due to insufficient knowledge of their clinical features. Here, we report two Chinese families with multiple AML cases carrying germline CEBPA mutations, one of which had 11 cases spanning four consecutive generations. Additionally, we collected clinical data of 57 AML patients from 22 families with germline CEBPA mutations, with 58.3% of them harboring double CEBPA mutations. The first mutation frequently occurred at the N-terminal of CEBP/α (78.6%), resulting in an exclusive expression of p30 of CEBPA (CEBPAp30). The second mutation was mostly found at the C-terminal of CEBP/α (CEBPAothers). Germline CEBPAp30 carriers had higher incidences of AML (80.36% vs. 42.86%) and earlier onset of AML (18 vs. 38.5 years old) compared to germline CEBPAothers carriers. Despite the high rates of relapse, most familial AML cases exhibited favorable overall survival (OS), with germline CEBPAp30 carriers having better survival outcomes (>25 years vs. 11 years for CEBPAothers carriers). Among the 27 healthy germline CEBPA-mutated carriers, we detected a pre-leukemia clone harboring a pathogenic IDH2 variant (R140Q)in one individual. These findings should aid in the genetic counseling and management of AML patients and healthy carriers with germline CEBPA mutations.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , Leukemia, Myeloid, Acute , Humans , Adult , CCAAT-Enhancer-Binding Protein-alpha/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/pathology , CCAAT-Enhancer-Binding Proteins/genetics , Mutation , Germ Cells/pathology , Prognosis
2.
J Leukoc Biol ; 114(5): 443-458, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37490847

ABSTRACT

This study tested the hypothesis that recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances polymorphonuclear neutrophils (PMNs) via interleukin (IL)-1ß to improve the prognosis of secondary infection in sepsis. The latter stage of sepsis is prone to induce immunosuppression, resulting in secondary fatal infections. Recombinant GM-CSF has become a way for sepsis-induced immunosuppression due to its immunomodulatory effect. However, the functional impact of GM-CSF on PMNs in sepsis remains obscure. This study aimed to study the role of recombinant GM-CSF on the bactericidal ability of PMNs in septic mice, assessing its effect on the prognosis of secondary pneumonia, and explore the mechanism of recombinant GM-CSF by intervening PMNs in patients with sepsis. The C57BL/6J sepsis mouse model was induced by cecal ligation and puncture. Recombinant murine GM-CSF (rmGM-CSF) was used in vivo when mice developed immunosuppression, which was characterized by abnormal bactericidal function of PMNs in peripheral blood. rmGM-CSF improved the prognosis of secondary pneumonia and reversed the function of PMNs. PMNs isolated by Percoll from septic patients were treated by recombinant human GM-CSF (rhGM-CSF) in vitro. The expression of CD11b, reactive oxygen species, phagocytosis, and neutrophil extracellular trap release in PMNs were enhanced by rhGM-CSF treatments. Whole-transcriptomic sequencing of mouse PMNs indicated that recombinant GM-CSF increased the expression of Il1b gene in PMNs. Blocking and inhibiting IL-1ß release effectively counteracted the enhancing effect of GM-CSF on the bactericidal function of PMNs. rmGM-CSF enhances the bactericidal function of PMNs in vivo and improves the prognosis of secondary pneumonia in septic mice, and recombinant GM-CSF increases IL-1ß precursor reserves, which, if stimulated, can rapidly enhance the bactericidal capacity of PMNs.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Sepsis , Humans , Mice , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Neutrophils/metabolism , Pseudomonas aeruginosa , Granulocyte Colony-Stimulating Factor/metabolism , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Sepsis/drug therapy , Prognosis
3.
Medicine (Baltimore) ; 102(17): e33098, 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37115091

ABSTRACT

Primary malignant lymphoma of the parotid gland is a rare entity. The disease is often misdiagnosed, and its survival factors remain unclear. This study included patients diagnosed with primary B-cell non-Hodgkin lymphoma of the parotid gland from 1987 to 2016 in the surveillance, epidemiology, and end results program. Univariate survival analysis was conducted using the Kaplan-Meier method, and multivariate analysis was performed using the Cox proportional hazards regression model. A competing risks regression model was applied to estimate the specific risks associated with parotid lymphoma mortality. A total of 1443 patients were identified. The overall survival of indolent primary B-cell lymphoma of the parotid gland was higher than that of aggressive lymphoma (hazard ratio 0.53, 95% confidence interval 0.44-0.64, P < .001), and older patients (≥70 years) exhibited inferior overall survival. Histological subtype and age are important prognostic factors in patients with primary B-cell non-Hodgkin lymphoma of the parotid gland.


Subject(s)
Lymphoma, B-Cell , Lymphoma , Parotid Neoplasms , Humans , Lymphoma/epidemiology , Parotid Gland , Parotid Neoplasms/epidemiology , SEER Program , Lymphoma, B-Cell/epidemiology , Prognosis
4.
Genomics ; 115(2): 110578, 2023 03.
Article in English | MEDLINE | ID: mdl-36775210

ABSTRACT

DEAH-box helicase 15 (DHX15) is ATP-dependent RNA helicase which is known for its role in RNA metabolism. Recent studies reported DHX15 involves in the intestinal immunity. However, the role of DHX15 (or RNA helicase) in intestinal development is poorly understood. Here, we revealed an unidentified role for dhx15 in regulating zebrafish intestinal development. We found the profound intestinal defects in dhx15 knockout zebrafish. Decreased proliferation and increased apoptosis of the intestine cells were observed when dhx15 were deleted. Further RNA genome wide analysis and qRT-PCR analysis showed the Wnt signaling pathway is down-regulated in the dhx15 knockout zebrafish. Thus, we concluded that dhx15 regulates zebrafish intestinal development through the Wnt signaling pathway. Here, we provided new insights into the role of dhx15 in intestinal development beyond its well-characterized role in intestinal immunity.


Subject(s)
Wnt Signaling Pathway , Zebrafish , Animals , RNA/metabolism , RNA Helicases/genetics , Zebrafish/genetics
5.
Front Immunol ; 13: 1029356, 2022.
Article in English | MEDLINE | ID: mdl-36389708

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system with no cure yet. Here, we report genetic engineering of hematopoietic stem cells (HSCs) to express myelin oligodendrocyte glycoprotein (MOG), specifically in platelets, as a means of intervention to induce immune tolerance in experimental autoimmune encephalomyelitis (EAE), the mouse model of MS. The platelet-specific αIIb promoter was used to drive either a full-length or truncated MOG expression cassette. Platelet-MOG expression was introduced by lentivirus transduction of HSCs followed by transplantation. MOG protein was detected on the cell surface of platelets only in full-length MOG-transduced recipients, but MOG was detected in transmembrane-domain-less MOG1-157-transduced platelets intracellularly. We found that targeting MOG expression to platelets could prevent EAE development and attenuate disease severity, including the loss of bladder control in transduced recipients. Elimination of the transmembrane domains of MOG significantly enhanced the clinical efficacy in preventing the onset and development of the disease and induced CD4+Foxp3+ Treg cells in the EAE model. Together, our data demonstrated that targeting transmembrane domain-deleted MOG expression to platelets is an effective strategy to induce immune tolerance in EAE, which could be a promising approach for the treatment of patients with MS autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Myelin-Oligodendrocyte Glycoprotein , Immune Tolerance , Central Nervous System
6.
Cancer Cell Int ; 22(1): 92, 2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35193582

ABSTRACT

BACKGROUND: DHX15 is one of the RNA helicase family members involving in several biological processes. Studies have reported that overexpression of DHX15 is related to cancer progression. However, the role of DHX15 in Burkitt lymphoma (BL) and latent Epstein-Barr virus (EBV) infection remains to be elucidated. METHODS: Expression of DHX15 was measured in BL patient by immunohistochemical staining. In vitro study, a CCK-8 assay was used to analyze cell proliferation and flow cytometry was performed to assess cell cycle, apoptosis and mitochondria membrane potential. Members of NF-κB signaling pathway and apoptotic-related proteins expression were measured by western-blot. EBV latent infection products and RNA polymerase III transcripts expression were determined by quantitative real-time PCR and western-blot. In vivo study, HE, IHC, TUNEL and ISH assays were used to analyze the effect of DHX15 on subcutaneous tumor nodes formation. RESULTS: DHX15 was overexpressed in Burkitt lymphoma patients and tends to be associated with poor progression-free survival and poor overall survival. Knockdown of DHX15 significantly inhibited BL tumor growth, reduced cell proliferation, induced cell cycle arrest and increased cell apoptosis. Further analysis showed that canonical NF-κB signaling and its downstream targets, mitochondria and Caspase were involved in the increased cell apoptosis after DHX15 gene knockdown. Furthermore, knockdown of DHX15 reduced EBV latent infection products expression and inhibited RNA polymerase III activity. CONCLUSION: DHX15 may be an oncogene in the development of BL and a potential therapeutic target for the treatment of BL and latent EBV infection.

7.
Cancer Sci ; 112(9): 3884-3894, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34077586

ABSTRACT

Gene alterations are recognized as important events in acute myeloid leukemia (AML) progression. Studies on hematopoiesis of altered genes contribute to a better understanding on their roles in AML progression. Our previous work reported a DEAH box helicase 15 (DHX15) R222G mutation in AML patients, and we showed DHX15 overexpression is associated with poor prognosis in AML patients. In this work, we further study the role of dhx15 in zebrafish developmental hematopoiesis by generating dhx15-/- zebrafish using transcription activator-like effector nuclease technology. Whole-mount in situ hybridization (WISH) analysis showed hematopoietic stem/progenitor cells were dramatically perturbed when dhx15 was deleted. Immunofluorescence staining indicated inhibited hematopoietic stem/progenitor cell (HSPC) proliferation instead of accelerated apoptosis were detected in dhx15-/- zebrafish. Furthermore, our data showed that HSPC defect is mediated through the unfolded protein response (UPR) pathway. DHX15 R222G mutation, a recurrent mutation identified in AML patients, displayed a compromised function in restoring HSPC failure in dhx15-/- ; Tg (hsp: DHX15 R222G) zebrafish. Collectively, this work revealed a vital role of dhx15 in the maintenance of definitive hematopoiesis in zebrafish through the unfolded protein respone pathway. The study of DHX15 and DHX15 R222G mutation could hold clinical significance for evaluating prognosis of AML patients with aberrant DHX15 expression.


Subject(s)
DEAD-box RNA Helicases/metabolism , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Unfolded Protein Response/genetics , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animals , Animals, Genetically Modified , Apoptosis/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Humans , In Situ Hybridization , Leukemia, Myeloid, Acute/metabolism , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
8.
Blood Adv ; 5(5): 1224-1238, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33646304

ABSTRACT

Gene therapy may lead to a cure for hemophilia B (HB) if it is successful. Data from clinical trials using adeno-associated virus (AAV)-mediated liver-targeted FIX gene therapy are very encouraging. However, this protocol can be applied only to adults who do not have liver disease or anti-AAV antibodies, which occur in 30% to 50% of individuals. Thus, developing a protocol that can be applied to all HB patients is desired. Our previous studies have demonstrated that lentivirus-mediated platelet-specific FIX (2bF9) gene therapy can rescue bleeding diathesis and induce immune tolerance in FIXnull mice, but FIX expression was only ∼2% to 3% in whole blood. To improve the efficacy, we used a codon-optimized hyperfunctional FIX-Padua (2bCoF9R338L) to replace the 2bF9 cassette, resulting in 70% to 122% (35.08-60.77 mU/108 platelets) activity levels in 2bCoF9R338L-transduced FIXnull mice. Importantly, sustained hyperfunctional platelet-FIX expression was achieved in all 2bCoF9R338L-transduced highly immunized recipients with activity levels of 18.00 ± 9.11 and 9.36 ± 12.23 mU/108 platelets in the groups treated with 11 Gy and 6.6 Gy, respectively. The anti-FIX antibody titers declined with time, and immune tolerance was established after 2bCoF9R338L gene therapy. We found that incorporating the proteasome inhibitor bortezomib into preconditioning can help eliminate anti-FIX antibodies. The bleeding phenotype in 2bCoF9R338L-transduced recipients was completely rescued in a tail bleeding test and a needle-induced knee joint injury model once inhibitors dropped to undetectable. The hemostatic efficacy in 2bCoF9R338L-transduced recipients was further confirmed by ROTEM and thrombin generation assay (TGA). Together, our studies suggest that 2bCoF9R338L gene therapy can be a promising protocol for all HB patients, including patients with inhibitors.


Subject(s)
Hemophilia B , Animals , Blood Platelets , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Hemophilia B/genetics , Hemophilia B/therapy , Mice
9.
Front Immunol ; 11: 964, 2020.
Article in English | MEDLINE | ID: mdl-32595633

ABSTRACT

Platelets are small anucleated blood components primarily described as playing a fundamental role in hemostasis and thrombosis. Over the last decades, increasing evidence has demonstrated the role of platelets in modulating inflammatory reactions and immune responses. Platelets harbor several specialized organelles: granules, endosomes, lysosomes, and mitochondria that can synthesize proteins with pre-stored mRNAs when needed. While the functions of platelets in the immune response are well-recognized, little is known about the potential role of platelets in immune tolerance. Recent studies demonstrate that platelet-specific FVIII gene therapy can restore hemostasis and induce immune tolerance in hemophilia A mice, even mice with preexisting anti-FVIII immunity. Here, we review the potential mechanisms by which platelet-targeted FVIII gene therapy restores hemostasis in the presence of anti-FVIII inhibitory antibodies and induces immune tolerance in hemophilia A.


Subject(s)
Antibodies/blood , Blood Platelets/metabolism , Factor VIII/genetics , Gene Targeting , Genetic Therapy , Hemophilia A/therapy , Hemostasis , Immune Tolerance , Animals , Antibodies/immunology , Blood Platelets/immunology , Factor VIII/immunology , Factor VIII/metabolism , Hemophilia A/blood , Hemophilia A/genetics , Hemophilia A/immunology , Humans , Treatment Outcome
10.
Blood Adv ; 3(20): 3099-3110, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31648333

ABSTRACT

The development of neutralizing anti-FVIII antibodies (inhibitors) is a major complication of FVIII protein replacement therapy in patients with hemophilia A (HA). Although multiple lines of evidence indicate that the immune response against FVIII is CD4 T-cell-dependent and many FVIII-derived CD4 epitopes have already been discovered, the role of T follicular helper (TFH) cells in FVIII inhibitor development is unknown. TFH cells, a newly identified subset of CD4 T cells, are characterized by expression of the B-cell follicle-homing receptor CXCR5 and PD-1. In this study, we show for the first time that IV FVIII immunization induces activation and accumulation and/or expansion of PD-1+CXCR5+ TFH cells in the spleen of FVIII-deficient (FVIIInull) mice. FVIII inhibitor-producing mice showed increased germinal center (GC) formation and increased GC TFH cells in response to FVIII immunization. Emergence of TFH cells correlated with titers of anti-FVIII inhibitors. Rechallenge with FVIII antigen elicited recall responses of TFH cells. In vitro FVIII restimulation resulted in antigen-specific proliferation of splenic CD4+ T cells from FVIII-primed FVIIInull mice, and the proliferating cells expressed the TFH hallmark transcription factor BCL6. CXCR5+/+ TFH-cell-specific deletion impaired anti-FVIII inhibitor production, confirming the essential role of CXCR5+/+ TFH cells for the generation of FVIII-neutralizing antibodies. Together, our results demonstrate that the induction of activated TFH cells in FVIIInull mice is critical for FVIII inhibitor development, suggesting that inhibition of FVIII-specific TFH-cell activation may be a promising strategy for preventing anti-FVIII inhibitor formation in patients with HA.


Subject(s)
Antibodies, Neutralizing/immunology , Factor VIII/immunology , Hemophilia A/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Factor VIII/genetics , Factor VIII/therapeutic use , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Hemophilia A/drug therapy , Hemophilia A/genetics , Hemophilia A/metabolism , Immunization , Immunophenotyping , Lymphocyte Depletion , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism
11.
Blood Adv ; 3(18): 2700-2711, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31515232

ABSTRACT

Gene therapy offers the potential to cure hemophilia A (HA). We have shown that hematopoietic stem cell (HSC)-based platelet-specific factor VIII (FVIII) (2bF8) gene therapy can produce therapeutic protein and induce antigen-specific immune tolerance in HA mice, even in the presence of inhibitory antibodies. For HSC-based gene therapy, traditional preconditioning using cytotoxic chemotherapy or total body irradiation (TBI) has been required. The potential toxicity associated with TBI or chemotherapy is a deterrent that may prevent patients with HA, a nonmalignant disease, from agreeing to such a protocol. Here, we describe targeted nongenotoxic preconditioning for 2bF8 gene therapy utilizing a hematopoietic cell-specific antibody-drug conjugate (ADC), which consists of saporin conjugated to CD45.2- and CD117-targeting antibodies. We found that a combination of CD45.2- and CD117-targeting ADC preconditioning was effective for engrafting 2bF8-transduced HSCs and was favorable for platelet lineage reconstitution. Two thirds of HA mice that received 2bF8 lentivirus-transduced HSCs under (CD45.2+CD117)-targeting ADC conditioning maintained sustained therapeutic levels of platelet FVIII expression. When CD8-targeting ADC was supplemented, chimerism and platelet FVIII expression were significantly increased, with long-term sustained platelet FVIII expression in all primary and secondary recipients. Importantly, immune tolerance was induced and hemostasis was restored in a tail-bleeding test, and joint bleeding also was effectively prevented in a needle-induced knee joint injury model in HA mice after 2bF8 gene therapy. In summary, we show for the first time efficient engraftment of gene-modified HSCs without genotoxic conditioning. The combined cocktail ADC-mediated hematopoietic cell-targeted nongenotoxic preconditioning that we developed is highly effective and favorable for platelet-specific gene therapy in HA mice.


Subject(s)
Blood Platelets/metabolism , Genetic Therapy/methods , Hemophilia A/drug therapy , Immunoconjugates/therapeutic use , Animals , Humans , Immunoconjugates/pharmacology , Male , Mice
12.
Asian Pac J Cancer Prev ; 20(3): 775-781, 2019 Mar 26.
Article in English | MEDLINE | ID: mdl-30909684

ABSTRACT

Background: The reason for the high incidence of gastric cancer (GC) in Xianyou County of China was largely unknown. We aimed to explore the potential sociodemographic risk factors and their associations to GC. Methods: A population-based case-control study was conducted during March 2013 and April 2016 in Xianyou County. All newly diagnosed patients of GC were recruited as cases, while controls were selected by matching for cases' sex, age (± 3 years) and the place of residence. Results: A total of 523 GC cases and 523 matched healthy controls were included in the final analysis with mean age of 66.27±8.81 years for cases and 66.31±8.83 years for controls, respectively. Participants with low socioeconomic status were observed with higher GC risk compared to those in high socioeconomic status (adjusted OR=2.10, 95% CI: 1.13-3.89). Compared to those regularly drink green tea, patients did not have this dietary habit had nearly 3-fold increased GC risk (adjusted OR=2.91, 95% CI: 1.38-6.13). Other dietary habit, including consumption of hard food, omission of breakfast, consumption of pickled vegetables 30 years ago, overeating were all associated with increased risk of GC. Interaction effect were found. Patients in low socioeconomic status and skipped breakfast had 10-fold higher risk of GC compared to reference group in high socioeconomic status and eat breakfast regularly (OR=10.71, 95% CI: 5.19-22.10). Furthermore, patients in low socioeconomic status and consumed pickled vegetable 30 years ago had 6-fold higher risk of GC compared to those in high socioeconomic status but did not intake pickled vegetables 30 years ago (OR=6.11, 95% CI: 3.87-9.66). Conclusion: High incidence of GC risk in Xianyou County might be partly attributed to various sociodemographic factors. Specific prevention effort could be target on population in low socioeconomic status combined with habit of breakfast omission or intake of pickled vegetables.


Subject(s)
Diet/adverse effects , Feeding Behavior , Stomach Neoplasms/diagnosis , Stomach Neoplasms/etiology , Aged , Breakfast , Case-Control Studies , China/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Life Style , Male , Middle Aged , Prognosis , Risk Factors , Stomach Neoplasms/epidemiology
13.
Medicine (Baltimore) ; 97(29): e11591, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30024563

ABSTRACT

BACKGROUND: BCL-2 Associated X (BAX) is an important modulator of apoptosis. The associations between BAX gene polymorphism and cancer susceptibility and prognosis in different ethnic groups and types of cancer have yielded controversial results. To reconcile the results, a systematic review followed by meta-analysis was performed to assess the associations. METHODS: A systematic search of Medline database (PubMed), EMBASE, China Biology Medicine disc, China National Knowledge Infrastructure, Wanfang databases for publications on BAX polymorphisms, and susceptibility and prognosis was carried out until July 2017. Retrieved 14 articles met the inclusions. Summary odds ratios (ORs) and hazard ratios (HRs) with their 95% confidence intervals (CIs) were harnessed to determine the strength of correlation between BAX polymorphisms and cancer susceptibility and prognosis, which were combined using fixed- or random-effects models as appropriate. RESULTS: A total of 12 trials involving 3321 cases and 3209 controls were included in our pooled analysis regarding the polymorphisms and the susceptibility of cancers. Overall, results of the present meta-analysis demonstrated that there was no significant association between BAX polymorphisms and susceptibility of cancers (OR = 1.052, 95% CI: 0.827-1.339, P = .679, A vs G). Even in a stratified analysis by ethnicity and the sources of control groups, the results were consistent. Four retrospective studies of 549 cases qualified for meta-analysis were identified to set forth the associations of the polymorphisms with cancer prognosis. Our results suggested that BAX gene polymorphisms were significantly associated with unfavorable prognosis (HR = 1.735, 95% CI: 1.368-2.202, P = .000, GG vs GA/AA). CONCLUSION: There is no significant association between BAX gene polymorphism and cancer susceptibility, but it probably contributes to increased adverse prognosis to cancer.


Subject(s)
Neoplasms/genetics , bcl-2-Associated X Protein/genetics , Genetic Predisposition to Disease , Humans , Polymorphism, Genetic , Prognosis
14.
J Cell Mol Med ; 22(5): 2612-2621, 2018 05.
Article in English | MEDLINE | ID: mdl-29512921

ABSTRACT

DHX15 plays a role in leukaemogenesis and leukaemia relapse. However, the mechanism underlying the transcriptional regulation of DHX15 in ALL has not been elucidated. Our present study aimed to explore the functional promoter region of DHX15 and to investigate the transcription factors controlling the transcription of this gene. A luciferase assay performed with several truncated constructs identified a 501-bp region as the core promoter region of DHX15. Site-directed mutagenesis, electrophoretic mobility shift and chromatin immunoprecipitation assays showed that ETS1 and SP1 occupied the DHX15 promoter. Furthermore, knockdown of ETS1 and SP1 resulted in suppression of DHX15, whereas the overexpression of these genes led to up-regulation of DHX15. Interestingly, in samples obtained from patients with ALL at diagnosis, both ETS1 and SP1 correlated positively with DHX15 expression. Additionally, differences in methylation of the DHX15 core promoter region were not observed between the patients and controls. In conclusion, we identified the core promoter region of DHX15 and demonstrated that ETS1 and SP1 regulated DHX15 expression in ALL.


Subject(s)
Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RNA Helicases/genetics , Sp1 Transcription Factor/metabolism , Base Pairing/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation/genetics , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-1/genetics , RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Initiation Site , Transcription, Genetic
15.
Oncotarget ; 8(52): 89643-89654, 2017 Oct 27.
Article in English | MEDLINE | ID: mdl-29163777

ABSTRACT

The role of DHX15, a newly identified DEAH-box RNA helicase, in leukemogenesis remains elusive. Here, we identified a recurrent mutation in DHX15 (NM_001358:c.664C>G: p.(R222G)) in one familial AML patient and 4/240 sporadic AML patients. Additionally, DHX15 was commonly overexpressed in AML patients and associated with poor overall survival (OS) (P=0.019) and relapse-free survival (RFS) (P=0.032). In addition, we found a distinct expression pattern of DHX15. DHX15 was highly expressed in hematopoietic stem cells and leukemia cells but was lowly expressed in mature blood cells. DHX15 was down-regulated when AML patients achieved disease remission or when leukemia cell lines were induced to differentiate. DHX15 silencing greatly inhibited leukemia cell proliferation and induced cell apoptosis and G1-phase arrest. In contrast, the restoration of DHX15 expression rescued cell viability and reduced cell apoptosis. In addition, we found that DHX15 was down-regulated when cell apoptosis was induced by ATO (arsenic trioxide); overexpression of DHX15 caused dramatic resistance to ATO-induced cell apoptosis, suggesting an important role for DHX15 in cell apoptosis. We further explored the mechanism of DHX15 in apoptosis and found that overexpression of DHX15 activated NF-kB transcription. Knockdown of DHX15 inhibited the nuclear translocation and activation of the NF-kB subunit P65 in leukemia cells. Several downstream targets of the NF-kB pathway were also down-regulated, and apoptosis-associated genes CASP3 and PARP were activated. In conclusion, this study represents the first demonstration that DHX15 plays an important role in leukemogenesis via the NF-kB signaling pathway and may serve as an independent prognostic marker for AML.

16.
Tumour Biol ; 39(7): 1010428317717107, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28670959

ABSTRACT

Xianyou county of Fujian province, located on the southeast coastal of China, has higher gastric cancer mortality. Chronic inflammation plays an important role in the occurrence of gastric cancer, in which the nuclear factor-κB signaling pathway of the inflammatory reaction begins and plays an important role in the amplification process. Studies have found that a single-nucleotide polymorphism of nuclear factor-κB signaling pathway molecules encoding genes is associated with gastric cancer, but the combined effect of the nuclear factor-κB signaling pathway gene has not been explained nor has been cardia and non-cardia gastric cancer risk factors and genetic susceptibility loci. New gastric cancer cases of the Fujian Xianyou Hospital were the research object. They were divided into cardia and non-cardia cancer in order to study a single-nucleotide polymorphism of the nuclear factor-κB signaling pathway important node molecules P50 and I kappa B encoding genes NFKB1 and NFKBIA by desorption ionization time of flight mass spectrometry analysis and by matrix-assisted laser mass spectrometry. The results showed that NFKB1 and NFKBIA single-nucleotide polymorphisms and gastric cancer are related and that the combined effects of polymorphisms in two genes and the NFKBIA gene monomer increased the risk of gastric cancer, and it was found that in different types of gastric cancer (the cardia and non-cardia cancer), susceptible polymorphism sites and combined effects are different.


Subject(s)
Genetic Association Studies , I-kappa B Proteins/genetics , NF-kappa B p50 Subunit/genetics , Stomach Neoplasms/genetics , China , Female , Genetic Predisposition to Disease , Genotype , Haplotypes/genetics , Humans , Male , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Polymorphism, Single Nucleotide , Risk Factors , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...