Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Am J Surg Pathol ; 45(10): 1391-1398, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34172624

ABSTRACT

Blast evaluation in patients with acute monocytic leukemias (AMoL) is notoriously difficult due to the lack of reliable surface markers and cytologic subtleties on the aspirate smears. While blasts of most nonmonocytic acute leukemias express CD34, available immunohistochemical antibodies to monocytic blasts also mark normal background mature monocytes. We searched for a potential biomarker candidate by surveying specific gene expression profiles of monocyte progenitors. Our investigations led us to IRF8, which is a lineage-specific transcription factor critical for the production of monocytic and dendritic cell progenitors. In this study, we tested and validated a monoclonal antibody to IRF8 as a novel immunohistochemical stain for trephine core biopsies of human bone marrow. We assessed the expression of IRF8 in 90 cases of AMoL, including posttherapy staging bone marrows, 23 cases of chronic myelomonocytic leukemia, 26 cases of other acute myeloid leukemia subtypes, and 18 normal control marrows. In AMoL, there was high correlation of IRF8-positive cells to aspirate blast count (R=0.95). Comparison of IRF8 staining to aspirate blast percentage in chronic myelomonocytic leukemia also showed good correlation (R=0.86). In contrast, IRF8-positive cells did not correlate with blast count in other subtypes of acute myeloid leukemia (R=0.56) and staining was <5% in all normal control marrows, even those with reactive monocytosis. We found that IRF8 was also weakly reactive in B cells and hematogones, with the latter accounting for rare cases of discrepancies. When IRF8 was used to categorize cases as AMoL, positive for residual leukemia or negative, the sensitivity was 98%, specificity was 82%, positive predictive value was 86%, and negative predictive value was 98%. These results demonstrate that IRF8 may serve as a clinically useful immunostain to diagnose and track AMoLs on bone marrow core biopsies. This can be particularly impactful in the setting of poor aspiration and focal blast increase. In the era of new targeted therapies that have been reported to induce monocytic outgrowths of leukemia, a marker for malignant monoblasts may prove even more critical.


Subject(s)
Biomarkers, Tumor/analysis , Immunohistochemistry , Interferon Regulatory Factors/analysis , Leukemia, Monocytic, Acute/metabolism , Monocyte-Macrophage Precursor Cells/chemistry , Aged , Biopsy , Bone Marrow Examination , Female , Humans , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Male , Middle Aged , Monocyte-Macrophage Precursor Cells/immunology , Monocyte-Macrophage Precursor Cells/pathology , Predictive Value of Tests , Proof of Concept Study , Reproducibility of Results
2.
J Immunother ; 44(4): 141-150, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33596023

ABSTRACT

Our previous research has shown that monocytic leukemia-associated antigen-34 (MLAA-34) was a novel antiapoptotic molecule with unique expression in acute monocytic leukemia (M5), making it an ideal target for T-cell-based immunotherapy. Here, we sought to identify HLA-A*0201-restricted cytotoxic T-lymphocyte (CTL) epitope of MLAA-34 by reverse immunology. In all, 10 HLA-A*0201 restricted epitopes of MLAA-34 were predicted by bioinformatics. MLAA-34324-332, MLAA-34293-301, and MLAA-34236-244 showed the strongest HLA-A*0201-binding affinity. The percentages of HLA-A*0201-MLAA-34236-244 tetramer+ CD8+ T cells in MLAA-34236-244-induced CTLs were raised apparently. Enzyme-linked immunospot showed that MLAA-34236-244 and MLAA-34324-332-specific CTLs produced a higher amount of interferon-γ. MLAA-34236-244-induced CTLs presented a stronger cytotoxic effect on THP-1 cells (HLA-A*0201+MLAA-34+) at various effector to target ratios. MLAA-34236-244 peptide vaccine could inhibit the tumor growth and improve mean survival time of leukemia-bearing human peripheral blood lymphocyte reconstituting severe combined immunodeficient mice. Mice immunized with MLAA-34236-244 vaccine had increased percentages of MLAA-34236-244 tetramer+ CD8+ T cells in the spleen after each round of immunization. High-purity CD8+ and CD4+ T cells were sorted by Dynabeads as effector cells. The killing activity of CD8+ T cells was higher than that of CD4+ T cells. CTLs derived from the MLAA-34 peptide vaccine group were significantly higher than other therapeutic groups and showed specific cytotoxicity to THP-1 cells. Increased interferon-γ and interleukin (IL)-2 and decreased IL-10 and IL-4 were seen in the MLAA-34236-244 peptide vaccine group. MLAA-34236-244 peptide (ILDRHNFAI) is an effective HLA-A*0201-restricted CTL epitope and that it may serve as a promising strategy in designing antigen-specific immunotherapy against MLAA-34-positive acute monocytic leukemia.


Subject(s)
Antigens, Neoplasm/immunology , Apoptosis Regulatory Proteins/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Leukemia, Monocytic, Acute/immunology , T-Lymphocytes, Cytotoxic/immunology , A549 Cells , Animals , Antineoplastic Agents , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Immunotherapy/methods , MCF-7 Cells , Mice , Mice, SCID , Peptides/immunology , THP-1 Cells/immunology , U937 Cells
4.
Comput Math Methods Med ; 2020: 8196147, 2020.
Article in English | MEDLINE | ID: mdl-32256683

ABSTRACT

Monocytic leukemia-associated antigen-42 (MLAA-42) is associated with excessive cell division and progression of leukemia. Thus, human MLAA-42 is considered as a promising target for designing of new lead molecules for leukemia treatment. Herein, the 3D model of the target was generated by homology modeling technique. The model was then evaluated using various cheminformatics servers. Moreover, the virtual screening studies were performed to explore the possible binding patterns of ligand molecules to MLAA's active site pocket. Thirteen ligand molecules from the ChemBank™ database were identified as they showed good binding affinities, scaffold diversity, and preferential ADME properties which may act as potent drug candidates against leukemia. The study provides the way to identify novel therapeutics with optimal efficacy, targeting MLAA-42.


Subject(s)
Antigens, Neoplasm/chemistry , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/immunology , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Amino Acid Sequence , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Computational Biology , Computer Simulation , Drug Design , Drug Screening Assays, Antitumor/statistics & numerical data , Humans , Leukemia, Monocytic, Acute/genetics , Ligands , Models, Molecular , Molecular Docking Simulation , Neoplasm Proteins/genetics , Protein Conformation , Protein Structure, Secondary , Structural Homology, Protein , User-Computer Interface
5.
Clin Lymphoma Myeloma Leuk ; 18(8): 541-547, 2018 08.
Article in English | MEDLINE | ID: mdl-29907544

ABSTRACT

BACKGROUND: The significance of FMS-like tyrosine kinase 3 (FLT3)-ITD mutation in acute myeloid leukemia (AML) prognosis has been well established. The aims of this study were to investigate the prognostic impact of the FLT3 protein (CD135) expression and its association with FLT3-ITD mutation, and to identify its role in minimal residual disease. PATIENTS AND METHODS: CD135 was measured by flow cytometry on leukemic blasts of 257 adults with de novo AML. High expression of CD135 ≥ 20% was correlated with clinical, laboratory, and other prognostic factors that influenced treatment outcome. FLT3-ITD mutation was tested by PCR. RESULTS: The frequency of CD135 expression was 138 (53.7%) of 257. FLT3-ITD was detected in (21.4%). Positive CD135 expression was associated with high total leukocyte count (P = .006), platelet count (P = .003), monocytic leukemia (P < .001), and CD34 (P = .008) and CD117 (P = .006) expression. CD135 expression ≥ 25% was a predictor of FLT3-ITD mutation (P = .03). CD135 overexpression was a negative predictor of complete remission and of postinduction minimal residual disease at days 14 and 28 (P < .001). CD135 had an adverse impact on overall and disease-free survival (68.5% vs. 15%, P = .002). Multivariate analysis indicated CD135 was the sole independent prognostic factor for overall survival (hazard ratio = 2.49; 95% confidence interval, 1.855-3.345; P < .001). CONCLUSION: CD135 is emerging as a prognostic factor, a new marker for minimal residual disease, and a potential novel therapeutic target of AML.


Subject(s)
Biomarkers, Tumor/analysis , Flow Cytometry , Leukemia, Monocytic, Acute/immunology , Lymphoid Progenitor Cells/immunology , fms-Like Tyrosine Kinase 3/analysis , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Disease-Free Survival , Egypt , Female , Genetic Predisposition to Disease , Humans , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/mortality , Lymphoid Progenitor Cells/drug effects , Male , Middle Aged , Mutation , Neoplasm, Residual , Phenotype , Predictive Value of Tests , Prospective Studies , Retrospective Studies , Time Factors , Young Adult , fms-Like Tyrosine Kinase 3/genetics
6.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(5): 1300-1306, 2017 Oct.
Article in Chinese | MEDLINE | ID: mdl-29070099

ABSTRACT

OBJECTIVE: To study the expression of stromal cell derived factor-1α (SDF-1α) receptor CXCR7 in acute monocytic leukemia (AML-M5), and its effects on proliferation, apoptosis, invasion of acute monocytic leukemia cell line THP-1. METHODS: CXCR7 protein and mRNA expression levels in THP-1 cells and peripheral blood mononuclear cells (PBMNC) from the newly diagnosed AML-M5 patients and normal individuals were detected by flow cytometry, Western blot and RT-PCR respectively. CCK8, Annexin V/PI double staining and Transwell assay were used to observe the effects of CXCR7 on the proliferation, apoptosis, and invasion of THP-1 cells in vitro. RESULTS: The expression of CXCR7 on immature cell surface of the newly diagnosed AML-M5 patients was significantly higher than that in the control group (P<0.05). CXCR7 was also highly expressed on THP-1 cells surface. The CXCR7 protein and mRNA levels in THP-1 cells and PBMNC of AML-M5 patients were significantly higher than those in the control group (P<0.05). The THP-1 cell proliferation activity was higher in SDF-1α-treated group, but this activity could be inhibited by CXCR7 antibody (P<0.01). CXCR7 antibody did not affect THP-1 cell apoptosis (P>0.05). CXCR7 antibody could inhibit SDF-1α -induced THP-1 cell invasiveness (P<0.01). CONCLUSION: CXCR7 highly expresses in AML-M5 patients and THP-1 cells, and involves in cell proliferation and invasion. The blocking CXCR7 expression can reduce the risk of AML-M5 cell infiltration.


Subject(s)
Leukemia, Monocytic, Acute/immunology , Receptors, CXCR/metabolism , THP-1 Cells/immunology , Apoptosis , Cell Proliferation , Chemokine CXCL12 , Humans , Leukocytes, Mononuclear , Receptors, CXCR/immunology , Receptors, CXCR4 , Signal Transduction
7.
Oncotarget ; 8(24): 39218-39229, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28424405

ABSTRACT

The Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has demonstrated promising efficacy in a variety of hematologic malignancies. However, the precise mechanism of action of the drug remains to be fully elucidated. Tumor-infiltrating macrophages presented in the tumor microenvironment have been shown to promote development and progression of B-cell lymphomas through crosstalk mediated by secreted cytokines and chemokines. Because Btk has been implicated in Toll-like receptor (TLR) signaling pathways that regulate macrophage activation and production of proinflammatory cytokines, we investigated the immunomodulatory effects of Btk inhibitor on macrophages. Our results demonstrate that Btk inhibition efficiently suppresses production of CXCL12, CXCL13, CCL19, and VEGF by macrophages. Furthermore, attenuated secretion of homeostatic chemokines from Btk inhibitor-treated macrophages significantly compromise adhesion, invasion, and migration of lymphoid malignant cells and even those not driven by Btk expression. The supernatants from Btk inhibitor-treated macrophages also impair the ability of endothelial cells to undergo angiogenic tube formation. Mechanistic analysis revealed that Btk inhibitors treatment downregulates secretion of homeostatic chemokines and cytokines through inactivation of Btk signaling and the downstream transcription factors, NF-κB, STAT3, and AP-1. Taken together, these results suggest that the encouraging therapeutic efficacy of Btk inhibitor may be due to both direct cytotoxic effects on malignant B cells and immunomodulatory effects on macrophages present in the tumor microenvironment. This novel mechanism of action suggests that, in addition to B-cell lymphomas, Btk inhibitor may also have therapeutic value in lymphatic malignancies and solid tumors lacking Btk expression.


Subject(s)
Leukemia, Monocytic, Acute/immunology , Lymphoma, B-Cell/immunology , Lymphoma, T-Cell/immunology , Macrophages/immunology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation , Chemokines/metabolism , Cytokines/metabolism , Humans , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/metabolism , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/metabolism , Macrophages/drug effects , Macrophages/metabolism , Piperidines , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tumor Cells, Cultured
8.
FEBS J ; 284(4): 568-585, 2017 02.
Article in English | MEDLINE | ID: mdl-28052562

ABSTRACT

Monocyte-to-macrophage differentiation is tightly controlled in vivo, as disruption of the normal differentiation program can lead to diverse disorders. Caspase-1, the first identified member of the caspase family, regulates differentiation in various cell types such as Th17 cells and adipocytes. However, the contribution of caspase-1 in monocyte-macrophage differentiation remains elusive. Here we report that caspase-1 is significantly downregulated in leukemia cells from patients with acute monocytic leukemia. By using the phorbol 12-myristate 13-acetate-induced cell differentiation model, we found that caspase-1 activation was required for the differentiation of human monocytes to macrophages. Further analysis of peroxisome proliferator-activated receptor γ (PPARγ) protein levels revealed that the monocyte-macrophage differentiation program could be divided into two stages. Caspase-1-mediated downregulation of PPARγ was important in the late stage of monocyte-macrophage differentiation; however, PPARγ protein levels had little effect on the early stage differentiation. Accumulation of PPARγ protein by troglitazone treatment potently suppressed the late stage of macrophage differentiation, which might be linked to inhibition of nuclear factor-κB activity. The data provide a plausible mechanistic basis by which caspase-1 promotes the differentiation of macrophages from monocytes.


Subject(s)
Caspase 1/genetics , Macrophages/immunology , Monocytes/immunology , NF-kappa B/genetics , PPAR gamma/genetics , Binding Sites , Caspase 1/immunology , Cell Differentiation/drug effects , Chromans/pharmacology , Gene Expression Regulation , Humans , Hypoglycemic Agents/pharmacology , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Macrophages/cytology , Macrophages/drug effects , Monocytes/drug effects , Monocytes/pathology , NF-kappa B/immunology , PPAR gamma/immunology , Primary Cell Culture , Protein Binding , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Thiazolidinediones/pharmacology , Troglitazone
9.
Genet Mol Res ; 14(2): 5630-41, 2015 May 25.
Article in English | MEDLINE | ID: mdl-26125761

ABSTRACT

Leukemia stem cells (LSCs) are regarded as the origin of leukemia and its recurrence. Side population (SP) cells possess some intrinsic stem cell properties and contain numerous LSCs. In this study, we examined the prognostic significance of cluster differentiation 47 (CD47) and identified the appropriate target for eliminating LSCs. We determined the percentage of SP cells in a THP-1 cell population and analyzed CD47 expression in different cell subsets. We then explored whether CD47 affected the phagocytic ability of macrophages to LSCs in vitro. Finally, the effect of anti-CD47 monoclonal antibodies, alone or combination with cytarabine, against leukemic cells was evaluated in vitro and in vivo to identify the optimal targets for the treatment of leukemia. We observed an SP sub-fraction at low frequency (1.81 ± 0.99%), which was a likely candidate for LSC enrichment. CD47 was more highly expressed on THP-1 LSCs (P < 0.05) and was an independent predictor of survival and refractory disease in THP-1-engrafted mice. Furthermore, the anti-CD47 monoclonal antibody stimulated preferential phagocytosis of LSCs by macrophages in vitro. Finally, single or combination treatment of THP-1 LSC-engrafted mice with cytarabine and anti-CD47 antibody resulted in targeting of LSCs and depletion of leukemia cells. These findings suggest that CD47 is an antibody target in LSCs and combination treatment with cytarabine and anti-CD47 monoclonal antibody represents an attractive option for the therapeutic targeting of acute monocytic leukemia.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD47 Antigen/immunology , Leukemia, Monocytic, Acute/drug therapy , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation/drug effects , Cell Line, Tumor , Combined Modality Therapy , Cytarabine/administration & dosage , Humans , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Macrophages/drug effects , Mice , Neoplastic Stem Cells/drug effects , Phagocytosis , Prognosis , Xenograft Model Antitumor Assays
10.
J Clin Pathol ; 68(4): 301-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25609576

ABSTRACT

AIMS: Pure erythroid leukaemia (PEL) is a rare subtype of acute myeloid leukaemia (AML) and its clinicopathological features are not well-defined. The aim of this study was to describe the immunophenotypic, cytogenetic and clinical features of PEL and to compare these with cases of AML with ≥ 50% erythroblasts. METHODS: Cases of PEL according to WHO morphological criteria diagnosed at three institutions from 1997 to 2013 were included. A comparison cohort comprised of AML with ≥ 50% erythroblasts. The clinical, histopathology, immunophenotypic and cytogenetic features of cases were analysed. We also reviewed the existing literature on PEL, and combined our cohort with previously reported cases of PEL in a pooled analysis. RESULTS: There were seven cases of PEL diagnosed at our institutions. There was a high incidence of either prior chemoradiotherapy exposure or evolution from pre-existing myelodysplastic syndrome (MDS) (71%). The leukaemic blasts frequently expressed glycophorin C (100%), CD117 (83%) and were myeloperoxidase negative (83%). Complex karyotypes were present in 83% of cases. Median overall survival was 2.9 months. Compared with AML with ≥ 50% erythroblasts, cases of PEL demonstrated a higher incidence of adverse-risk cytogenetics (p=0.01) and prior exposure to chemoradiotherapy (p=0.01). CONCLUSIONS: PEL appears to be a unique entity that is often secondary or therapy related, commonly features a complex karyotype and has a poor prognosis. It is morphologically and immunophenotypically distinct from other cases of AML with erythroid hyperplasia.


Subject(s)
Erythroblasts , Leukemia, Monocytic, Acute/diagnosis , Leukemia, Monocytic, Acute/epidemiology , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Cell Proliferation , Erythroblasts/chemistry , Erythroblasts/immunology , Erythroblasts/pathology , Genetic Predisposition to Disease , Humans , Hyperplasia , Immunophenotyping , Karyotype , Karyotyping , Leukemia, Monocytic, Acute/classification , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Leukemia, Monocytic, Acute/therapy , Male , Middle Aged , Phenotype , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Factors , Victoria/epidemiology , Young Adult
11.
Fitoterapia ; 92: 85-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24185011

ABSTRACT

The potential effects of globularifolin, an acylated iridoid glucoside, on cell survival, inflammation markers and free radicals scavenging were investigated. Viability assay on human myelomomonocytic cell line THP-1 and human peripheral blood mononuclear cells (PBMC) using the Cell-Titer Blue assay proved that globularifolin had no toxic effect at the tested concentrations. Conversely, it is proportional to the dose globularifolin increased growth of THP-1 cells (p <0.01). On human PBMC, globularifolin at 6.25 and 12.5 µM concentrations showed a stimulatory effect, while at 12.5-200 µM it suppressed response of PBMC to stimulation with phytohemagglutinin (PHA). Globularifolin (50-200 µM) enhanced neopterin formation dose-dependently, whereas tryptophan breakdown was not influenced. At 50-200 µM in unstimulated PBMC in THP-1 cells, globularifolin induced a significant expression of nuclear factor-κB (NF-κB) as was quantified by Quanti-Blue assay. By contrast, in lipopolysaccharide (LPS)-stimulated cells, the higher concentrations of globularifolin suppressed NF-κB expression dose-dependently and a significant decrease was observed at 200 µM concentration. A positive correlation was found between increased neopterin and NF-κB activity (p <0.01). Similarly, a positive correlation was observed between neopterin levels in mitogen-induced cells and NF-κB activity in LPS-stimulated cells after treatment with globularifolin (p=0.001). The free radical scavenging capacity of globularifolin evaluated by Oxygen Radical Absorbance Capacity (ORAC) assay showed relative ORAC values of 0.36±0.05 µmol Trolox equivalent/µmol. All together, results show that natural antioxidant globularifolin might represent a potential immunomodulatory as well as proliferative agent, which deserves further in vitro and in vivo studies.


Subject(s)
Antioxidants/pharmacology , Free Radicals/metabolism , Iridoid Glucosides/pharmacology , Leukocytes, Mononuclear/drug effects , NF-kappa B/metabolism , Neopterin/biosynthesis , Tryptophan/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Immunologic Factors/pharmacology , Inflammation Mediators/metabolism , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides , Mitogens/metabolism , Phytotherapy , Plant Extracts/pharmacology , Plantago/chemistry
12.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(3): 596-600, 2013 Jun.
Article in Chinese | MEDLINE | ID: mdl-23815905

ABSTRACT

This study was aimed to investigate the clinical features of CD56(+) patients with acute monocytic leukemia (AML-M5) and their prognostic significance. The data of 76 newly-diagnosed patients from our hospital were analyzed retrospectively. Patients were divided into two groups: CD56(+) group (21 patients) and CD56(-) group (55 patients). The clinical features, CR rate, relapse rate, the duration of CR, and survival time of patients between the two groups were compared. The results indicated that the CD56(+) antigen was observed in 21 patients (27.6%), their median age was 51.5 years and with a range 16 - 70 years. Of the 21 CD56(+) patients, the high WBC count was found in 57.1% CD56(+) patients (12/21), but it only in 15% CD56(-) patients (P < 0.05). The extramedullary infiltration was seen in 13 CD56(+) patients, and accounted for 62% (13/21), meanwhile this infiltration was found in 18 CD56(-) patients (18/55) and accounted for 33% (P < 0.05). All cases immunophenotypically highly expressed CD13, CD33, CD64, CD11b, cMPO, CD38, in which only the expression frequency of CD11b was positively related with CD56 (r = 0.59, P < 0.05). The CR rate in CD56(+) group accounted for 60.0%, and had no significant difference in comparison with that in CD56(-) group. In CD56(+) group the relapse rate was 75% (P = 0.042), the mean duration of CR was 5.5 months (95%CI, 3.1 - 8.6, P = 0.002), the median overall survival time was 10.1 months (95%CI, 2.3 - 16.3, P = 0.001). and all these had statistical significance as compared with that in CD56(-) group. It is concluded that CD56(+) AML-M5 patients always complicate with high WBC count and extramedullary infiltration, their CR rate and duration of CR are lower and shorter respectively, their relapse rate and prognosis are high and poor respectively.


Subject(s)
Leukemia, Monocytic, Acute/diagnosis , Leukemia, Monocytic, Acute/immunology , Adolescent , Adult , Aged , CD56 Antigen/metabolism , Female , Humans , Immunophenotyping , Male , Middle Aged , Prognosis , Young Adult
13.
Neurobiol Aging ; 34(5): 1389-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23273574

ABSTRACT

A mutation in the human cystatin C gene leads to familial cerebral amyloid angiopathy. This disease is known as "hereditary cerebral hemorrhage with amyloidosis-Icelandic type" or "hereditary cystatin C amyloid angiopathy." The mutant cystatin C protein forms aggregates and amyloid, within the central nervous system almost exclusively in connection with the vascular system. It was not known whether immune cells could remove mutant cystatin C protein aggregates. Ex vivo mutant cystatin C protein aggregates, both in solution and dried onto a glass surface, induced adhesion to the substrate, differentiated the THP-1 monocyte cell line and led to a proinflammatory response. Aggregates were also taken up by both THP-1 cells and THP-1 derived macrophages. These are the same responses induced by other amyloidogenic protein species, such as amyloid ß protein and amylin, supporting the model of all amyloidogenic proteins being toxic due to common structural motifs. Proinflammatory response induced by the ex vivo mutant cystatin C protein aggregates suggests that vascular inflammation plays an important role in hereditary cerebral hemorrhage with amyloidosis-Icelandic type. Ex vivo protein aggregates of cystatin C might better model cellular behavior than in vitro-generated aggregates or supplement in vitro material.


Subject(s)
Cystatin C/immunology , Cystatin C/pharmacology , Endocytosis/immunology , Inflammation/immunology , Leukemia, Monocytic, Acute/immunology , Cell Adhesion , Cell Differentiation/drug effects , Cell Line, Tumor , Cerebral Amyloid Angiopathy , Cystatin C/genetics , Endocytosis/drug effects , Genetic Variation/genetics , Humans
15.
Rinsho Ketsueki ; 53(11): 1932-6, 2012 Nov.
Article in Japanese | MEDLINE | ID: mdl-23257676

ABSTRACT

A 6-year-old boy was diagnosed with FLT3-ITD+acute monoblastic leukemia (AMoL). He showed resistance to 2 cycles of induction chemotherapy with etoposide, cytarabine, and mitoxantrone or idarubicin performed according to the Japan Pediatric Leukemia/Lymphoma Study Group (JPLSG) AML-05 protocol. His condition was also refractory to salvage FLAI-GO (fludarabine, cytarabine, idarubicin, and gemtuzumab ozogamicin) chemotherapy. Sequential administration of sorafenib at doses of up to 300 mg/day resulted in the first remission. He underwent bone marrow transplantation from his HLA 2-locus mismatched father. Recurrence was observed on post-transplantation day 71. A sustained partial response was observed after alternate-day readministration of sorafenib 150mg/day. In spite of a donor lymphocyte infusion, his blast cell count increased on day 245. Chemotherapy with an increased dose of sorafenib reduced the blast cell count. Although a second HLA-mismatched allogeneic peripheral blood stem cell transfusion was performed, the patient died of regimen-related toxicity. Herein, we report a pediatric case of primary refractory FLT3-ITD+ AMoL. Further prospective studies are necessary to validate the efficacy of sorafenib treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Monocytic, Acute/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Bone Marrow Transplantation/adverse effects , Child , Fatal Outcome , Humans , Inverted Repeat Sequences , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/therapy , Male , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Sorafenib , fms-Like Tyrosine Kinase 3/analysis
16.
Thromb Res ; 130(4): e229-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22940059

ABSTRACT

Our previous study has demonstrated that the Toll-like receptor 4 (TLR4) signaling pathways contribute to the induction of tissue factor (TF) expression by anti-ß(2)-glycoprotein I/ß(2)-glycoprotein I (anti-ß(2)GPI/ß(2)GPI) in human acute monocytic leukemia cell line THP-1. In this study, we focused on the identification of the downstream targets of the TLR4 pathways. When THP-1 cells were treated with anti-ß(2)GPI/ß(2)GPI complex, enhanced TF expression was observed, along with induced phosphorylation of p38, ERK1/2 and JNK1/2 MAPKs. When the activity of MAPKs was blocked by their corresponding inhibitors (SB203580: p38; U0126: ERK; SP600125: JNK), the expression of TF was reduced significantly. Furthermore, the anti-ß(2)GPI/ß(2)GPI-induced phosphorylation of p38, ERK1/2 and JNK1/2 was inhibited significantly by TAK-242, a blocker of the signaling transduction mediated by the intracellular domain of TLR4; sc-204013, a specific inhibitor of IRAKs, was also able to partially inhibit the phosphorylation of the MAPKs. Our results demonstrated that MAPKs (p38, ERK1/2 and JNK1/2) were the crucial downstream targets of the anti-ß(2)GPI/ß(2)GPI-triggered TLR4 signaling pathways in THP-1 cells. This essential role of MAPKs may also promote better understanding of the pathogenesis of antiphospholipid syndrome (APS).


Subject(s)
Antigen-Antibody Complex/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Leukemia, Monocytic, Acute/immunology , MAP Kinase Signaling System , Thromboplastin/genetics , Toll-Like Receptor 4/immunology , beta 2-Glycoprotein I/immunology , Antiphospholipid Syndrome/genetics , Antiphospholipid Syndrome/immunology , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Leukemic , Humans , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Leukemia, Monocytic, Acute/genetics , MAP Kinase Signaling System/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 20(4): 857-62, 2012 Aug.
Article in Chinese | MEDLINE | ID: mdl-22931642

ABSTRACT

This study was purposed to analyse the immunophenotypic characteristics of chronic myelomonocytic leukemia (CMML), myelodysplastic syndromes (MDS) and acute monocytic leukemia (AML-M5b) by using multiparameter flow cytometry, and to explore its significance in diagnosis and differential diagnosis. The immunophenotypic characteristics of bone marrow samples from 14 CMML patients, 48 MDS patients, 46 AML-M5b patients and 18 normal persons were analyzed and compared by multiparametric flow cytometry. The results showed that the ratio of monocytes in CMML patients was obviously higher than that in MDS, AML-M5b patients and normal persons (P < 0.05), but there was no statistically significant difference between bone marrow samples of MDS and AML-M5b patients as well as normal persons. The ratio of blast cells in MDS patients was obviously higher than that in normal persons (P < 0.05), but did not show significant difference as compared with CMML patients. The ratio of mature granulocytes in AML-M5b patients was obviously lower than that in CMML and MDS patients as well as normal person bone marrow (P < 0.05). Certain differences of CD45/SSC characteristics in MDS, AML-M5b and CMML patients were found in comparison with normal persons. The abnormal expression of CD2, CD56, and CD14 tailing phenomenon were observed in CMML patients in comparison with bone marrow samples of MDS, AML-M5b and normal persons (P < 0.05). Lack and decrease of CD15 expression in MDS and CMML patients was significant different from AML-M5b and normal persons marrow, abnormal expression rate of CD15 in CMML patients was higher than that in MDS patients (P < 0.05), the CD13/CD11b/CD16 abnormal expression of granulocytes was seen in both CMML and MDS patients, but there was no statistically significant difference between them. Other antigens showed abnormality of varying degrees, but did not have any statistical significance. It is concluded that MDS, CMML and AML-M5b displayed a certain degree of similarity, and also possess their own immunophenotype characteristics. Comprehensive analysis of immunophenotype by multiparameter flow cytometry may be important for differential diagnosis among CMML, MDS and AML-M5b. High percentage of monocytes, abnormal coexpression of CD2, CD56 and CD14 tailing phenomenon, lack or decrease of CD15 as well as abnormal expression of CD13/CD11b/CD16 in granulocytes may play important roles in diagnosis of CMML.


Subject(s)
Flow Cytometry/methods , Leukemia, Monocytic, Acute/diagnosis , Leukemia, Myelomonocytic, Chronic/diagnosis , Myelodysplastic Syndromes/diagnosis , Case-Control Studies , Humans , Immunophenotyping/methods , Leukemia, Monocytic, Acute/immunology , Leukemia, Myelomonocytic, Chronic/immunology , Myelodysplastic Syndromes/immunology
18.
J Immunol ; 187(12): 6227-34, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22084431

ABSTRACT

Resting human NK cells require a two-stage activation process that we have previously described as "priming" and "triggering." NK-sensitive tumor cells provide both priming and triggering signals. NK-resistant tumors evade lysis, mostly by failure to prime; however, we recently reported a tumor cell line (CTV-1) that primes resting NK cells but fails to trigger lysis. In this article, we report two additional leukemia cell lines that prime NK cells but are resistant to lysis. Tumor-mediated NK priming is via CD2 binding to a ligand within CD15 on the tumor cell. NK-resistant RAJI cells became susceptible to NK lysis following transfection and expression of CD15. Blockade of CD15 on K562 cells or on CD15(+) RAJI cells significantly inhibited lysis, as did blockade of CD2 on resting NK cells. NK priming via CD2 induced CD16 shedding, releasing CD3ζ to the CD2, leading to its phosphorylation and the subsequent phosphorylation of linker for activation of T cells and STAT-5 and synthesis of IFN-γ. Blockade of C-type lectin receptors significantly suppressed the tumor-mediated priming of NK cells, whereas blockade of Ig-superfamily-like receptors had no effect at the NK-priming stage. Tumor priming of resting NK cells was irrespective of HLA expression, and blockade of HLA-killer Ig-like receptor interactions did not influence the incidence or degree of priming. However, CD15-CD2 interactions were critical for NK priming and were required, even in the absence of HLA-mediated NK inhibition. Tumor-mediated priming led to a sustained primed state, and the activated NK cells retained the ability to lyse NK-resistant tumors, even after cryopreservation.


Subject(s)
CD2 Antigens/metabolism , Cytotoxicity, Immunologic , Fucosyltransferases/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lewis X Antigen/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, KIR/physiology , Resting Phase, Cell Cycle/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Lineage/immunology , Coculture Techniques , Disease Resistance , Fucosyltransferases/metabolism , Humans , Killer Cells, Natural/cytology , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/metabolism , Leukemia, Monocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Acute/metabolism , Leukemia, Myelomonocytic, Acute/pathology , Lewis X Antigen/metabolism , Ligands , Lymphocyte Activation/immunology , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/immunology , Signal Transduction/immunology
19.
Molecules ; 16(6): 4836-49, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21666550

ABSTRACT

Many diseases occur when the immune system is weakened. Intracellular signals activate immuno-responsive cells to produce cytokines that modulate the immune response. Schisandra chinensis has been used traditionally to treat general fatigue, neurasthenia, and spontaneous sweating. In the present study, the effect of constituents of S. chinensis on cytokine release by human monocytic leukemia cells (THP-1) was tested using microparticle-based flow cytometric analysis. Two major lignans, schizandrin (Sch) and gomisin A (Gom A), were identified and shown to induce interleukin (IL)-8, macrophage inflammatory protein-1ß (MIP-1ß), and granulocyte-macrophage-colony stimulating factor (GM-CSF) release by THP-1 cells. By reverse transcription polymerase chain reaction (RT-PCR) or quantitative real-time PCR, there was a dose-dependent increase of IL-8, MIP-1ß and GM-CSF mRNA levels. Thus, Sch and Gom A from S. chinensis enhance cytokine release by THP-1 cells and this effect occurs through mRNA upregulation. Upregulation of MIP-1ß and GM-CSF in particular may have clinical applications. Therefore, S. chinensis may be therapeutically beneficial by promoting humoral and cell-mediated immune responses.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Immunologic Factors/pharmacology , Schisandra/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Drugs, Chinese Herbal/chemistry , Humans , Immunologic Factors/chemistry , Leukemia, Monocytic, Acute/immunology , Models, Biological , RNA, Messenger/metabolism
20.
J Immunol ; 186(5): 3265-76, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21282517

ABSTRACT

mAbs that recognize peptides presented on the cell surface by MHC class I molecules are potential therapeutic agents for cancer therapy. We have previously demonstrated that these Abs, which we termed TCR mimic mAbs (TCRm), reduce tumor growth in models of breast carcinoma. However, mechanisms of TCRm-mediated tumor growth reduction remain largely unknown. In this study, we report that these Abs, in contrast to several mAbs used currently in the clinic, destroy tumor cells independently of immune effector mechanisms such as Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). We found that TCRm-mediated apoptosis of tumor cells was associated with selective and specific binding of these Abs to peptide/HLA class I complexes, which triggered the activation of JNK and intrinsic caspase pathways. This signaling was accompanied by the release of mitochondrial cytochrome c and apoptosis-inducing factor. TCRm-induced apoptosis in tumor cells was completely inhibited by soluble MHC tetramers loaded with relevant peptide as well as with inhibitors for JNK and caspases. Furthermore, mAbs targeting MHC class I, independent of the peptide bound by HLA, did not stimulate apoptosis, suggesting that the Ab-binding site on the MHC/peptide complex determines cytotoxicity. This study suggests the existence of mechanisms, in addition to ADCC and CDC, through which these therapeutic Abs destroy tumor cells. These mechanisms would appear to be of particular importance in severely immunocompromised patients with advanced neoplastic disease, since immune cell-mediated killing of tumor cells through ADCC and CDC is substantially limited in these individuals.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Apoptosis/immunology , Molecular Mimicry/immunology , Receptors, Antigen, T-Cell/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/metabolism , Cell Line, Tumor , Female , HLA-A2 Antigen/metabolism , Humans , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Leukemia, Monocytic, Acute/therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Nude , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...