Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Clin Lab ; 70(10)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39382918

ABSTRACT

BACKGROUND: The aim of this study was to investigate the changes of T lymphocyte subsets (Th1, Th2, Tc1, Tc2, and Th17) and memory T lymphocyte subsets (Tcm and Tem) in patients with multiple myeloma (MM) at different stages of the disease. METHODS: In total, 25 newly diagnosed patients with MM were selected as the study subjects and 30 healthy people were selected as the control group. The subsets of T lymphocytes such as Th1, Th2, Tc1, Tc2, Th17, Tcm, and Tem in the peripheral blood were detected by flow cytometry at the time of initial diagnosis, infection, and remission. RESULTS: Th1, Tem, and Tcm cells in MM patients showed a significant decrease compared to the control group. Th2 and Th17 cells in MM patients showed a significant increase compared to the control group. Total Th1 cells and memory Th1 cells in MM patients with bacterial infection were significantly higher than at initial diagnosis (p < 0.05). The Tcm of Th2 cells in the remission stage were significantly higher than those in MM patients with no remission. CONCLUSIONS: MM patients have decreased Th1 cells and increased Th2 and Th17 cells. The changes in memory Th1 cells were related to bacterial infection in MM patients. The increase of Tcm of Th2 cells may be associated with disease remission. The balance of T lymphocyte subsets plays an important role in the pathogenic course of MM.


Subject(s)
Multiple Myeloma , T-Lymphocyte Subsets , Humans , Multiple Myeloma/immunology , Multiple Myeloma/blood , Multiple Myeloma/diagnosis , Male , Middle Aged , Female , Aged , T-Lymphocyte Subsets/immunology , Flow Cytometry , Adult , Memory T Cells/immunology , Case-Control Studies , Th2 Cells/immunology , Th1 Cells/immunology , Th17 Cells/immunology
2.
Clin Lab ; 70(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39257117

ABSTRACT

BACKGROUND: This study aimed to explore the changes in Th cells and cytokines in the peripheral blood of patients with multiple myeloma before and after treatment and at the time of the bacterial infection. METHODS: In total, 23 newly diagnosed MM patients admitted to the Hospital and 23 healthy individuals were selected as the study group and the control group, respectively. Flow cytometry was used to detect the Th1 and Th2 lymphocytes and cytokines, such as IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, INF-γ, IL-17A, IL-1b, TNF-α, TNF-ß, and IL-12P70, in the peripheral blood of the patients at initial diagnosis, before and after treatment, and at the time of the bacterial infection. RESULTS: The Th1% and Th1/Th2 ratio at the time of the initial diagnosis were lower in the MM patients than in the control group, whereas the Th2% at initial diagnosis was higher in the MM patients than in the control group. The levels of IL-6, IL-8, IL-10, and IL-17A at initial diagnosis were higher in the MM patients than in the control group. After 4 cycles of treatment, the Th2% in the patients was lower than before the treatment and the Th1/Th2 ratio in the patients was higher than before the treatment. The Th1% and the levels of IL-6, IL-8, IL-10, and INF-γ increased, while the level of IL-12P70 decreased, when MM patients got a bacterial infection. The abovementioned differences were statistically significant (p < 0.05). CONCLUSIONS: The Th1/Th2 deviation affects the immune function of the MM patients. There were significant changes in the Th1 and Th2 lymphocytes and cytokines in newly diagnosed MM patients after the treatment. The changes in the Th lymphocytes and cytokines may be an indicator of bacterial infection.


Subject(s)
Cytokines , Multiple Myeloma , Th1 Cells , Th2 Cells , Humans , Multiple Myeloma/blood , Multiple Myeloma/immunology , Cytokines/blood , Male , Female , Middle Aged , Th1 Cells/immunology , Th2 Cells/immunology , Aged , Th1-Th2 Balance , Adult , Case-Control Studies , Bacterial Infections/blood , Bacterial Infections/immunology , Bacterial Infections/diagnosis
3.
Clin Lab ; 70(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38469780

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) with t(8;21)(q22;q22.1); RUNX1::RUNX1T1 has a relatively favorable prognosis with a high complete remission rate and long disease-free survival. METHODS AND RESULTS: Here we describe a patient who had AML with t(8;21)(q22;q22.1); RUNX1::RUNX1T1. Cooperating mutations including KRAS and ASXL1, and with other abnormal karyotype del(17) and with a myelomonocytic differentiation. CONCLUSIONS: The patient relapsed despite achieving a morphologic complete remission (CR).


Subject(s)
Leukemia, Myeloid, Acute , Translocation, Genetic , Humans , Core Binding Factor Alpha 2 Subunit/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Mutation
4.
Med Sci Monit ; 26: e920541, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32054823

ABSTRACT

BACKGROUND Multiple myeloma is featured by the proliferation of malignant plasma cell in bone marrow. We aimed to demonstrate the effects of valproic acid combined with GANT61 on multiple myeloma cell proliferation and clarify its mechanism. MATERIAL AND METHODS Multiple myeloma cells were exposed to valproic acid, GANT61, or the combination of valproic acid and GANT61, respectively. MTT assay was performed to detect the cell viability. Quantitative reverse transcriptase polymerase chain reaction and western blotting were used to detect mRNA and expression levels of proteins in Hedgehog signaling pathway. The Q-value of the combination regime was calculated to evaluate the drug combination effect. RESULTS Both valproic acid and GANT61 alone inhibited multiple myeloma cell proliferation in a dose-dependent manner compared to the control. In the presence of GANT61 or not, valproic acid inhibited multiple myeloma cell proliferation in a time-dependent manner. These 2 drugs had a synergistic effect at valproic acid concentration of ≥4 mM. Expression analysis showed that valproic acid significantly inhibited the expression levels of PTCH1, GLI1, and HES-1. GANT61 enhanced the inhibition of Hedgehog signaling pathway mediated by valproic acid. CONCLUSIONS GANT61 and valproic acid inhibited multiple myeloma cell proliferation synergistically by inhibiting the Hedgehog signaling pathway. The present study may provide a combination regime for the therapy of multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Valproic Acid/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Hedgehog Proteins/metabolism , Humans , Multiple Myeloma/pathology , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Valproic Acid/therapeutic use
6.
Leuk Res ; 138: 107455, 2024 03.
Article in English | MEDLINE | ID: mdl-38368721

ABSTRACT

OBJECTIVE: To explore the involvement of TFEB-mediated autophagy-lysosomal mechanisms in multiple myeloma (MM) during bortezomib treatment. METHODS: MM cells were exposed to bortezomib or subjected to TFEB knockdown. CCK assay was used to assess the cell proliferation. Western blotting and fluorescent staining were conducted to examine autophagy and lysosomes. The TFEB expression pattern was analyzed, and whole transcriptome sequencing was carried out. Additionally, TFEB target genes were predicted using the GTRD(http://gtrd.biouml.org/) website, and pathway analysis was performed. RESULTS: Bortezomib demonstrated a dose-dependent and time dependent inhibition of cell proliferation. In MM cells treated with bortezomib, LC3B, Beclin-1, TFEB, and Lamp1 exhibited upregulation in a time- and concentration-dependent manner. LysoTracker dye labeling showed an increase in lysosomes in the bortezomib-treated group. Moreover, bortezomib elevated the expression of lysosome-associated factor Lamp1. Bortezomib promoted the nuclear translocation of TFEB, leading to decreased cytoplasmic TFEB and increased nuclear TFEB. TFEB gene silencing reversed bortezomib's inhibitory effect on MM cell lines, significantly reducing autophagosome expression and lysosome numbers. Furthermore, bioinformatic analysis identified the MAPK pathway as a potential downstream target of TFEB. CONCLUSION: Bortezomib effectively inhibits MM cell proliferation and induces autophagy, partly through TFEB-mediated mechanisms, with potential involvement of the MAPK pathway.


Subject(s)
Multiple Myeloma , Humans , Bortezomib/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Autophagy , Autophagosomes/metabolism , Lysosomes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
7.
Indian J Pathol Microbiol ; 66(4): 865-867, 2023.
Article in English | MEDLINE | ID: mdl-38084551

ABSTRACT

In patients with acute myeloid leukemia (AML), about 25%-35% of patients have a history of other hematological diseases, 10% of patients have a history of malignant tumors in other systems and have received cytotoxic treatment including chemotherapy and/or radiation, and the disease is categorized as therapy-related acute myeloid leukemia (t-AML) according to the World Health Organization (WHO) classification of tumors of hematopoietic and lymphoid tissues. Two subsets of t-AML are generally recognized based on the nature of prior treatments and the characteristics of the disease. The most common type occurs after exposure to alkylating agents and/or radiation, with a latent period of 5 to 10 years. The less common type occurs after treatment with agents targeting topoisomerase II and has a shorter latent period of 1 to 5 years. The majority of these cases are associated with balanced recurrent chromosomal translocations frequently involving MLL at 11q23, RUNX1 at 21q22, or CBFB at 16q22 and morphologically resemble the features of de novo AML associated with these translocations. Here, we describe a rare case of a 48-year-old female with ovarian cancer who developed AML with CBFB/MYH11 fusion, less than two years after exposure to paclitaxel and carboplatin chemotherapy.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Ovarian Neoplasms , Humans , Female , Middle Aged , Leukemia, Myeloid, Acute/pathology , Translocation, Genetic , Antineoplastic Agents/adverse effects , Gene Rearrangement , Ovarian Neoplasms/drug therapy , Core Binding Factor beta Subunit/genetics , Myosin Heavy Chains
8.
J Biomol Struct Dyn ; 41(2): 525-537, 2023 02.
Article in English | MEDLINE | ID: mdl-34844510

ABSTRACT

Olmutinib, the third-generation tyrosine kinase inhibitor, is applied in treating non-small cell lung cancer (NSCLC). The aim of this study is to elucidate the interaction mechanism of olmutinib with human α-1 acid glycoprotein (HAG), an important carrier protein, by mean of multi-spectroscopic and molecular simulation techniques. Fluorescence spectral results confirmed that the fluorescence of this carrier protein can be quenched by olmutinib in the static quenching mode, and this anticancer drug possesses a moderate binding affinity on HAG. The evidence from thermodynamic analysis, replacement interaction with ANS and sucrose, and computational simulation results showed that hydrogen bonding, hydrophobic interactions, and van der Waals forces involved the olmutinib-HAG complexation process. The results from UV-vis, 3D fluorescence and synchronous fluorescence spectroscopy proved that binding anticancer drug olmutinib caused the alteration in the microenvironment around Trp residues. And, circular dichroism spectral results provided the support for the conformational alterations in the carrier protein. The data also proved that olmutinib preferably bound to the hydrophobic cavity of HAG and the binding distance between the two was 2.21 nm. In addition, it can be found that the presence of some metal ions such as Zn2+, Ca2+, Ni2+ and Cu2+ would exert a certain extent effect on the olmutinib-HAG complexation process.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Binding Sites , Protein Binding , Orosomucoid/metabolism , Molecular Docking Simulation , Spectrometry, Fluorescence/methods , Circular Dichroism , Thermodynamics , Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Tumor Microenvironment
9.
Chemosphere ; 345: 140530, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37890791

ABSTRACT

A template-free pyrolysis route has been developed using condensation-assembly precursors made of trimethoxyboroxane (TMB) and melamine (M) to cater the requirements of an industrial real-world environment. The precursors contain abundant B-N bonds and exhibit a high level of interconnectivity, resulting in 3D-PBN with enhanced mechanical properties and the ability to be easily customized in terms of shape. Moreover, 3D-PBN demonstrates rapid adsorption kinetics and excellent reusability, efficiently removing up to 270% of its own weight of fuel within 30 s and being readily regenerated through simple calcination. Even after undergoing 50 cycles, the mechanical properties remain at a remarkable 80%, while the adsorption performance exceed 95%. Furthermore, a comprehensive analysis of thermal behavior from precursor to 3D-PBN has been conducted, leading to the proposal of a molecular-scale evolution process comprising four major steps. This understanding enables us to control the phase reaction and regulate the composition of the products, which is crucial for determining the characteristics of the final product.


Subject(s)
Boron Compounds , Porosity , Boron Compounds/chemistry
10.
Cell Cycle ; 19(16): 2063-2073, 2020 08.
Article in English | MEDLINE | ID: mdl-32677544

ABSTRACT

PURPOSE: This study aimed to explore the effect of GANT61 on regulating cell proliferation, cell apoptosis and cell cycle, and to investigate whether GANT61 would function in multiple myeloma (MM) via inhibiting Notch pathway. Methods: RPMI-8226 and U266 cells were treated by GANT61 (0, 2.5, 5.0, 10.0, 20.0, 30.0, 40.0, 50.0 µmol/L) for 18, 24 and 36 hours (h), and cell proliferation was detected by Cell Counting Kit 8. Then these cells were treated by GANT61 at 0, 2.5, 5.0, 10.0 µmol/L for 24 h or treated by 10.0 µmol/L GANT61 for 0, 18, 24 and 36 h, and cell apoptosis rate, apoptosis markers and cell cycle were detected by AV/PI, Western blot, and PI staining. Notch1, Jagged1, Jagged2 and Hes1 expressions were detected by qPCR and Western blot. Further rescue experiments were conducted by upregulating Notch1. Results: In RPMI-8226 and U266 cells, GANT61 inhibited cell proliferation, increased cell apoptosis rate and cell percentage of G1/G0 phase while decreased cell percentage of S phase in a dose- and time-dependent manner. Besides, GANT61 inhibited Notch1, Jagged1, Jagged2 and Hes1 expressions in a dose- and time-dependent manner as well. In rescue experiments, Notch1 upregulation attenuated the inhibition of cell proliferation, promotion of cell apoptosis, induction of G1/G0 cycle retardation and repression of Notch signaling pathway induced by GANT61 treatment in RPMI-8226 and U266 cells. Conclusions: GANT61 suppresses cell proliferation, promotes cell apoptosis and induces G1/G0 cycle retardation with a dose- and time-dependent manner through inhibiting Notch pathway in MM. ABBREVIATIONS: MM: Multiple myeloma; Hh: Hedgehog; EMT: epithelial mesenchymal transition; AML: acute myeloid leukemia; GANT61: GLI antagonist; DMSO: dimethyl sulfoxide; CCK-8: Cell Counting Kit 8; C-Caspase 3: Cleaved Caspase 3; Bcl-2: B-cell lymphoma-2; RT-qPCR: real-time quantitative polymerase chain reaction; OD: optical density; PTCH1: Patched1.


Subject(s)
Apoptosis , G1 Phase Cell Cycle Checkpoints , Multiple Myeloma/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Notch/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Time Factors , Zinc Finger Protein GLI1/metabolism
11.
Biomed Pharmacother ; 127: 110167, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32344258

ABSTRACT

BACKGROUND: To investigate the effects of valproic acid (VPA) on autophagic flux in multiple myeloma (MM) cells. METHODS AND RESULTS: Cell proliferation was assayed by the Cell Counting Kit-8 assay. The qRT-PCR was used to measure the expressions of LC3-II at mRNA level. Autophagic flux was measured by LC3-II turnover using western blot analysis and flow cytometry using the fluorescent dye Cyto-ID. An assay using the RFP-GFP-LC3 tandem construct was performed to monitor autophagic flux. Cell proliferation assay showed that VPA could inhibit the proliferation of MM cells and the inhibitory effects were enhanced with the extension of time. The qRT-PCR and western blot showed that the expression level of LC3-II in the VPA plus CQ group was significantly higher than that in CQ group. Cyto-ID autophagy test showed that the intracellular average fluorescence intensity in VPA plus CQ group was significantly higher than that in control and VPA group (all p < 0.001). The results of RFP-GFP-LC3 tandem construct showed that the numbers of yellow puncta and red puncta in VPA group was higher than that in control group. CONCLUSIONS: VPA could inhibit the proliferation of MM cells and the inhibitory effects were enhanced with the extension of time. VPA could enhance autophagic flux in MM cells, and the increase of autophagosomes was caused by autophagy enhancement rather than inhibition. These findings provided rationale for the treatment of MM with VPA.


Subject(s)
Autophagy/drug effects , Cell Proliferation/drug effects , Multiple Myeloma/drug therapy , Valproic Acid/pharmacology , Antineoplastic Agents/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Microtubule-Associated Proteins/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology
12.
Anticancer Agents Med Chem ; 13(2): 270-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22934698

ABSTRACT

MicroRNAs (miRNAs) and promoter hypermethylation are vital epigenetic mechanisms for transcriptional inactivation of tumor suppressor. IRX1 is a newly identified tumor suppressor gene and hypermethylation involves the decreased expression in gastric cancer. However, the microRNA regulatory mechanism on IRX1 expression is still unclear. In this study, we report an IRX1-targeting miRNA-544, which directly targets 3'-UTR of IRX1 gene by luciferase reporter assay. miR-544 suppresses the protein expression of IRX1 gene by Western blot and immunocytochemistry. Ectopic expression of miR-544 promotes cell proliferation and cell cycle progression significantly in vitro on gastric cancer cells. The study suggests that miR-544 is an oncogenic microRNA in gastric cancer. Over expression of miR-544 contributes to the inactivation and low-expression of IRX1 in gastric cancer. These findings are helpful for clarifying the molecular mechanisms involved in gastric carcinogenesis and indicate that miR-544 is a key regulator in switching cell cycle on or off. miR-544 may be a potential molecular target in miRNA-based strategy on gastric cancer.


Subject(s)
MicroRNAs/genetics , Molecular Targeted Therapy , Stomach Neoplasms/genetics , Cell Cycle , Cell Line , Cell Proliferation , Gene Expression Regulation, Neoplastic/genetics , Humans , RNA, Neoplasm/genetics , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL