Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Diagnostics (Basel) ; 14(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786343

ABSTRACT

Circulating cfRNA in plasma has emerged as a fascinating area of research with potential applications in disease diagnosis, monitoring, and personalized medicine. Circulating RNA sequencing technology allows for the non-invasive collection of important information about the expression of target genes, eliminating the need for biopsies. This comprehensive review aims to provide a detailed overview of the current knowledge and advancements in the study of plasma cfRNA, focusing on its diverse landscape and biological functions, detection methods, its diagnostic and prognostic potential in various diseases, challenges, and future perspectives.

2.
Front Genet ; 14: 1266990, 2023.
Article in English | MEDLINE | ID: mdl-38046047

ABSTRACT

Introduction: Metagenomic next-generation sequencing (mNGS) has emerged as a powerful tool for rapid pathogen identification in clinical practice. However, the parameters used to interpret mNGS data, such as read count, genus rank, and coverage, lack explicit performance evaluation. In this study, the developed indicators as well as novel parameters were assessed for their performance in bacterium detection. Methods: We developed several relevant parameters, including 10M normalized reads, double-discard reads, Genus Rank Ratio, King Genus Rank Ratio, Genus Rank Ratio*Genus Rank, and King Genus Rank Ratio*Genus Rank. These parameters, together with frequently used read indicators including raw reads, reads per million mapped reads (RPM), transcript per kilobase per million mapped reads (TPM), Genus Rank, and coverage were analyzed for their diagnostic efficiency in bronchoalveolar lavage fluid (BALF), a common source for detecting eight bacterium pathogens: Acinetobacter baumannii, Klebsiella pneumoniae, Streptococcus pneumoniae, Staphylococcus aureus, Hemophilus influenzae, Stenotrophomonas maltophilia, Pseudomonas aeruginosa, and Aspergillus fumigatus. Results: The results demonstrated that these indicators exhibited good diagnostic efficacy for the eight pathogens. The AUC values of all indicators were almost greater than 0.9, and the corresponding sensitivity and specificity values were almost greater than 0.8, excepted coverage. The negative predictive value of all indicators was greater than 0.9. The results showed that the use of double-discarded reads, Genus Rank Ratio*Genus Rank, and King Genus Rank Ratio*Genus Rank exhibited better diagnostic efficiency than that of raw reads, RPM, TPM, and in Genus Rank. These parameters can serve as a reference for interpreting mNGS data of BALF. Moreover, precision filters integrating our novel parameters were built to detect the eight bacterium pathogens in BALF samples through machine learning. Summary: In this study, we developed a set of novel parameters for pathogen identification in clinical mNGS based on reads and ranking. These parameters were found to be more effective in diagnosing pathogens than traditional approaches. The findings provide valuable insights for improving the interpretation of mNGS reports in clinical settings, specifically in BALF analysis.

3.
Diagnostics (Basel) ; 12(4)2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35453875

ABSTRACT

We developed an artificial intelligence (AI) model that evaluates the feasibility of AI-assisted multiparameter flow cytometry (MFC) diagnosis of acute leukemia. Two hundred acute leukemia patients and 94 patients with cytopenia(s) or hematocytosis were selected to study the AI application in MFC diagnosis of acute leukemia. The kappa test analyzed the consistency of the diagnostic results and the immunophenotype of acute leukemia. Bland-Altman and Pearson analyses evaluated the consistency and correlation of the abnormal cell proportion between the AI and manual methods. The AI analysis time for each case (83.72 ± 23.90 s, mean ± SD) was significantly shorter than the average time for manual analysis (15.64 ± 7.16 min, mean ± SD). The total consistency of diagnostic results was 0.976 (kappa (κ) = 0.963). The Bland-Altman evaluation of the abnormal cell proportion between the AI analysis and manual analysis showed that the bias ± SD was 0.752 ± 6.646, and the 95% limit of agreement was from -12.775 to 13.779 (p = 0.1225). The total consistency of the AI immunophenotypic diagnosis and the manual results was 0.889 (kappa, 0.775). The consistency and speedup of the AI-assisted workflow indicate its promising clinical application.

4.
J Transl Med ; 19(1): 369, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446054

ABSTRACT

To evaluate whether low coverage whole genome sequencing is suitable for the detection of malignant pelvic mass and compare its diagnostic value with traditional tumor markers. We enrolled 63 patients with a pelvic mass suspicious for ovarian malignancy. Each patient underwent low coverage whole genome sequencing (LCWGS) and traditional tumor markers test. The pelvic masses were finally confirmed via pathological examination. The copy number variants (CNVs) of whole genome were detected and the Stouffers Z-scores for each CNV was extracted. The risk of malignancy (RM) of each suspicious sample was calculated based on the CNV counts and Z-scores, which was subsequently compared with ovarian cancer markers CA125 and HE4, and the risk of ovarian malignancy algorithm (ROMA). Receiver Operating Characteristic Curve (ROC) were used to access the diagnostic value of variables. As confirmed by pathological diagnosis, 44 (70%) patients with malignancy and 19 patients with benign mass were identified. Our results showed that CA125 and HE4, the CNV, the mean of Z-scores (Zmean), the max of Z-scores (Zmax), the RM and the ROMA were significantly different between patients with malignant and benign masses. The area under curve (AUC) of CA125, HE4, CNV, Zmax, and Zmean was 0.775, 0.866, 0.786, 0.685 and 0.725 respectively. ROMA and RM showed similar AUC (0.876 and 0.837), but differed in sensitivity and specificity. In the validation cohort, the AUC of RM was higher than traditional serum markers. In conclusion, we develop a LCWGS based method for the identification of pelvic mass of suspicious ovarian cancer. LCWGS shows accurate result and could be complementary with the existing diagnostic methods.


Subject(s)
Neoplasms, Glandular and Epithelial , Ovarian Neoplasms , Algorithms , Biomarkers, Tumor/genetics , CA-125 Antigen , Female , Humans , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Proteins , ROC Curve , WAP Four-Disulfide Core Domain Protein 2 , Whole Genome Sequencing
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(6): 499-506, 2020 Jun.
Article in Chinese | MEDLINE | ID: mdl-32696739

ABSTRACT

Objective To investigate the inhibitory effect of abnormal nuclear localization of the nuclear localization signal-retinoic acid receptor α (NLS-RARα) on cell differentiation and its mechanism of nuclear transport. Methods Over-expression of HA-NLS-RARα and empty vector in HEK293T cells and U937 cells were achieved through a lentivirus vector and were assigned as NLS-RARα over-expression (NR) group and negative control (NC) group. Extracted nucleoproteins and cytosolic proteins of NC and NR groups of HEK293T cells and U937 cells were detected by Western blot analysis in order to demonstrate the localization of NLS-RARα. Meanwhile, immunofluorescence assay was performed to explore the localization of NLS-RARα. The real-time quantitative PCR and Western blot analysis were used to detect difference in the mRNA and protein expression of CD11b and CEBPß in the NR cells treated with 1, 25-dihydroxyvitamin D3 (1, 25D3) compared with NC cells treated with 1, 25D3. Mass spectrometric analysis and co-immunoprecipitation were conducted to screen the transport proteins which were associated with NLS-RARα, which was followed by the verification of nuclear accumulation of NLS-RARα by the transfection of transport protein small interfering RNA. Results Western blot assay and immunofluorescence showed that NLS-RARα was mainly located in the nucleus. And the qRT-PCR analysis and western blot assay showed a significant decrease in the mRNA and protein expression of CD11b and CEBPß in the NR group compared with the NC group. It demonstrated that NLS-RARα inhibited cell differentiation. Mass spectrometric analysis and COIP demonstrated that KPNA2 (importin α1) and KPNB1 (importin ß1) interacted with NLS-RARα, and the knockdown of KPNA2/KPNB1 inhibited the nuclear accumulation of NLS-RARα. Conclusion Abnormal localization of NLS-RARα inhibits cell differentiation via binding to KPNA2 and KPNB1 into the nucleus.


Subject(s)
Cell Nucleus , Nuclear Localization Signals , Active Transport, Cell Nucleus , Cell Differentiation , Cell Nucleus/metabolism , HEK293 Cells , Humans , Protein Binding , Retinoic Acid Receptor alpha/metabolism , U937 Cells , alpha Karyopherins , beta Karyopherins
6.
Biochem Biophys Res Commun ; 528(2): 276-284, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32475642

ABSTRACT

A majority of acute promyelocytic leukaemia (APL) cases are characterized by the PML-RARα fusion gene. Previous studies have shown that neutrophil elastase (NE) can cleave PML-RARα and is important for the development of APL. Here, we demonstrate that one of the cleavage products of PML-RARα, NLS-RARα, can block cell differentiation by repressing the expression of the target genes within the retinoic acid signalling pathway. The results of reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot analysis showed that NLS-RARα depressed the expression of the cell differentiation marker protein, CD11b and CEBPß, as well as the retinoic acid signalling pathway target genes, RARß and CEBPε. Studies have shown that NLS-RARα forms heterodimers with retinoid X receptor α(RXRα) and interacts with SMRT. When treated with all-trans retinoic acid (ATRA), NLS-RARα exhibits diminished transcriptional activity compared to RARα. Moreover, in the presence of high doses of ATRA, NLS-RARα could be degraded along with the consequent transactivation of retinoic acid signalling pathway target genes and cell differentiation induction in a dose- and time-dependent manner. Together, these results indicate that NLS-RARα blocks cell differentiation by inhibiting the retinoic acid signalling pathway.


Subject(s)
Cell Differentiation , Nuclear Localization Signals/metabolism , Retinoic Acid Receptor alpha/chemistry , Retinoic Acid Receptor alpha/metabolism , Signal Transduction , Tretinoin/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Gene Expression Regulation/drug effects , Humans , Models, Biological , Nuclear Receptor Co-Repressor 2/metabolism , Protein Binding/drug effects , Proteolysis/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Tretinoin/pharmacology
7.
Cell Signal ; 69: 109567, 2020 05.
Article in English | MEDLINE | ID: mdl-32036017

ABSTRACT

The promyelocytic leukemia-retinoic acid receptor α (PML/RARα) is hypothesized to play a vital role in the pathogenesis of acute promyelocytic leukemia (APL). A previous study has demonstrated that PML/RARα is cleaved by neutrophil elastase (NE) in early myeloid cells, which leads to an increase in the nuclear localization signal (NLS) in RARα and in the incidence of APL. In this study, we explored the effects of NLS-RARα on acute myeloid leukemia (AML) cells and studied the mechanism of its localization. LV-NLS-RARα recombinant lentivirus and negative control LV-NC lentivirus were transfected into HL-60 cells and U937 cells while mutant NLS-RARα were transfected into U937 cells, and all groups were treated with 1α, 25-dihydroxyvitamin D3(1,25D3). The results showed that NLS-RARα was located mainly in the nucleus while mutant NLS-RARα was located in the cytoplasm. Overexpression of NLS-RARα downregulated the expression of CD11b, CD11c, CD14, and three forms of CEBPß compared to the overexpression of NC and mutant NLS-RARα. It was speculated that the abnormal localization of NLS-RARα was mediated via importin-α/ß in the pathogenesis of APL. By producing point mutations in the two NLSs in NLS-RARα, we showed that the nuclear import of NLS-RARα was mainly dependent on the NLS of the RARα portion. Subsequently, we found that importin-α1 (KPNA2)/importin-ß1 (KPNB1) participates in the nuclear transport of NLS-RARα. Taken together, abnormal localization of NLS-RARα blocks the differentiation of APL cells, and nuclear localization of NLS-RARα depends on NLS of the RARα portion and is mediated via binding with importin-α/ß.


Subject(s)
Cell Nucleus/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Localization Signals/physiology , Retinoic Acid Receptor alpha/physiology , alpha Karyopherins/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , HL-60 Cells , Humans , U937 Cells
8.
Cell Signal ; 65: 109431, 2020 01.
Article in English | MEDLINE | ID: mdl-31654721

ABSTRACT

The fusion oncogene, promyelocytic leukemia (PML)-retinoic acid receptor-α (RARα), is crucial for acute promyelocytic leukemia (APL) pathogenesis. Previous studies have reported that PML-RARα is cleaved by neutrophil elastase (NE), an early myeloid-specific serine protease, leading to translocation of the nuclear localization signal (NLS) of the PML protein to the N-terminal of RARα. This study was designed to evaluate the value of NLS-RARα in the early diagnosis of APL. To investigate the potential functional role of NLS-RARα in leukemogenesis, HL-60 and U937 cell lines were transfected with NLS-RARα lentivirus and negative control (LVNC). The results showed that the induced expression of NLS-RARα down-regulated expressions of CD11b, CD11c, and CD14 compared to the LVNC group induced by 1α, 25-dihydroxyvitamin D3(1,25(OH)2D3). This suggested that NLS-RARα overexpression inhibited granulocytic and monocytic differentiation of myeloid leukemia cells. In addition, Wright-Giemsa staining, flow cytometry, respiratory burst assay, and NBT reduction assay all confirmed the importance of NLS-RARα in differentiation. The mechanistic investigations revealed that induced NLS-RARα expression inhibited 1,25(OH)2D3-induced granulocytic differentiation by regulating the cell cycle regulators p19INK4D, p21WAF1/CIP1, cyclinD1, cyclin E1, and pRB. Furthermore, the cleaved protein NLS-RARα enhanced the oncogenicity of U937 cells in NOD/SCID mice. These findings collectively demonstrated that NLS-RARα blocked granulocytic and monocytic differentiation of myeloid leukemia cells by inhibiting the downstream targets of the RARα signal pathway and the cell cycle. This may provide a promising new target and method for diagnosing and treating APL.


Subject(s)
Carcinogenesis/pathology , Cell Differentiation , Leukemia, Promyelocytic, Acute/pathology , Nuclear Localization Signals/metabolism , Retinoic Acid Receptor alpha/chemistry , Retinoic Acid Receptor alpha/metabolism , Animals , Calcitriol/pharmacology , Carcinogenesis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Humans , Mice, Inbred NOD , Mice, SCID , Structure-Activity Relationship
9.
Cell Signal ; 60: 57-64, 2019 08.
Article in English | MEDLINE | ID: mdl-30965092

ABSTRACT

MicroRNAs have been shown to be involved in various cell processes, including proliferation, apoptosis and differentiation. However, little is known about their function in granulopoiesis. In the present study, overexpression and knockdown experiments revealed that miR-15b was required to block the proliferation of NB4 and HL60 cells and induce them differentiated to granulocyte lineage. Moreover, we identified CCNE1 as a direct target of miR-15b, and demonstrated that CCNE1 was involved in cell differentiation and proliferation in acute promyelocytic leukemia cells. In addition, we demonstrated a novel pathway in which miR-15b regulated cells arrested in the G0/G1 phase and promoted terminal differentiation of cells by targeting CCNE1, which could modulate the cell cycle effort pRb in APL cells. These events blocked cell proliferation and promoted granulocyte differentiation. In conclusion, our data highlighted, for the first time, the important role of miR-15b in myeloid differentiation and suggested the potential role of miR-15b in cancer therapy.


Subject(s)
Cyclin E/metabolism , Leukemia, Promyelocytic, Acute/metabolism , MicroRNAs/physiology , Oncogene Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , HL-60 Cells , Humans
10.
Oncol Res ; 27(7): 809-818, 2019 Jul 12.
Article in English | MEDLINE | ID: mdl-30982491

ABSTRACT

RAS-responsive element-binding protein 1 (RREB1) is a transcription factor that is implicated in RAS signaling and multiple tumors. However, the role of RREB1 in acute myeloid leukemia has not been studied. We found that RREB1 is overexpressed in AML patients and myeloid leukemia cell lines (NB4 and HL-60), and RREB1 expression was significantly decreased during granulocytic differentiation of myeloid leukemia cells induced by all-trans retinoic acid (ATRA). Then we performed a RREB1 knockdown assay in NB4 and HL-60 cells; the results showed that knockdown of RREB1 upregulated expression of CD11b, CEBPß, and microRNA-145 (miR-145), which hinted that knockdown of RREB1 enhanced granulocytic differentiation of myeloid leukemia cells. In addition, inhibitor of miR-145 can offset the enhanced effect on granulocytic differentiation mediated by downregulation of RREB1. These collective findings demonstrated that RREB1 blocks granulocytic differentiation of myeloid leukemia cells by inhibiting the expression of miR-145 and downstream targets of the RAS signal pathway. These may provide a promising therapeutic target for AML patients.


Subject(s)
DNA-Binding Proteins/metabolism , Granulocytes/pathology , Leukemia, Myeloid, Acute/pathology , Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Granulocytes/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , MicroRNAs/genetics , Signal Transduction , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics , Tretinoin/pharmacology , ras Proteins/metabolism
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(12): 1082-1087, 2019 Dec.
Article in Chinese | MEDLINE | ID: mdl-31894006

ABSTRACT

Objective To investigate the effect of all-trans retinoic acid (ATRA) on cell differentiation in nuclear localization signal-retinoic acid receptor-α (NLS-RARα)-overexpressing leukemia cells. Methods Lentivirus was used to infect NB4 and U937 cells to overexpress NLS-RARα. Fluorescent microscope, real-time PCR and Western blot analysis were performed to measure the mRNA and protein expression of NLS-RARα, CD11b and CEBPß. Modified Wright staining was used to observe the cell morphology. Results NLS-RARα was overexpressed successfully after the lentivirus infection. Overexpressed NLS-RARα could repress the expression of CD11b and CEBPß. NLS-RARα could be degraded by ATRA in a concentration- and time-dependent manner; ATRA promoted cell differentiation in both negative control cells and NLS-RARα-overexpressing cells. Conclusion ATRA promotes the differentiation of human leukemia cells by degrading NLS-RARα protein.


Subject(s)
Cell Differentiation , Leukemia, Promyelocytic, Acute/pathology , Nuclear Localization Signals/metabolism , Retinoic Acid Receptor alpha/metabolism , Tretinoin/pharmacology , Humans , U937 Cells
12.
Cell Signal ; 54: 1-9, 2019 02.
Article in English | MEDLINE | ID: mdl-30453015

ABSTRACT

In acute promyelocytic leukemia (APL), all-trans retinoic acid (ATRA) treatment induces granulocytic differentiation and maturation. MicroRNAs play pivotal roles in formation of the leukemic phenotype. Previously, microRNA-382-5p (miR-382-5p) was upregulated in acute myeloid leukemia (AML) with t(15;17). In the present study, we found that miR-382-5p expression was elevated with ATRA-induced differentiation of APL. To investigate the potential functional role of miR-382-5p in APL differentiation, an APL cell line was transfected with miR-382-5p mimics, inhibitors, or negative control (NC). The results showed in APL cell line NB4 that miR-382-5p downregulation upon ATRA treatment was a key event in the drug response. Mechanistic investigations revealed that miR-382-5p targeted the ATRA-regulated tumor suppressor gene PTEN through direct binding to its 3' UTR. Enforced expression of miR-382-5p or specific PTEN inhibitors inhibited ATRA-induced granulocytic differentiation via regulation of the cell cycle regulator cyclinD1. Conversely, PTEN overexpression promoted differentiation and enhanced sensitivity of NB4 cell line to physiological levels of ATRA. Finally, we found that PTEN overexpression restored PML nuclear bodies (NBs). Taken together, these results demonstrated that up-regulated miR-382-5p in NB4 cell line inhibited granulocytic differentiation through the miR-382-5p/PTEN axis, uncovering PTEN as a critical element in the granulocytic differentiation program induced by ATRA in APL.


Subject(s)
Cyclin D1/metabolism , Leukemia, Promyelocytic, Acute , MicroRNAs/physiology , PTEN Phosphohydrolase/metabolism , Tretinoin/pharmacology , Antineoplastic Agents/pharmacology , Cell Differentiation , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , THP-1 Cells
13.
Cell Signal ; 53: 390-399, 2019 01.
Article in English | MEDLINE | ID: mdl-30448346

ABSTRACT

Actin-like 6A (ACTL6A), a component of BAF chromatin remodeling complexes, is important for cell differentiation. Nevertheless, its role and mechanism in acute promyelocytic leukemia (APL) has not been reported. To identify the genes that may participate in the development of APL, we analyzed data from an APL cDNA microarray (GSE12662) in the NCBI database, and found that ACTL6A was up-regulated in APL patients. Subsequently, we investigated the function and mechanisms of ACTL6A in myeloid cell development. The expression of ACTL6A was gradually decreased during granulocytic differentiation in all-trans retinoic acid-treated NB4 and HL-60 cells, and phorbol myristate acetate-treated HL-60 cells. We also found that knockdown of ACTL6A promoted differentiation in NB4 and HL-60 cells, and decreased the levels of Sox2 and Notch1. Mechanistically, ACTL6A interacted with and was co-localized with Sox2 and p53. Meanwhile, CBL0137, an activator of p53, decreased the expression of ACTL6A and promoted differentiation in NB4 and HL-60 cells. These findings suggest that the inhibition of ACTL6A promotes differentiation via the Sox2 and Notch1 signaling pathways. Furthermore, the differentiation promoted by inhibiting ACTL6A could be regulated by p53 via its physical interaction with ACTL6A.


Subject(s)
Actins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Receptor, Notch1/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , HL-60 Cells , Humans , Protein Interaction Maps , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL