Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Pediatr Blood Cancer ; 71(6): e30970, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38556751

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare hematologic neoplasm characterized by the clonal proliferation of Langerhans-like cells. Colony-stimulating factor 1 receptor (CSF1R) is a membrane-bound receptor that is highly expressed in LCH cells and tumor-associated macrophages. In this study, a soluble form of CSF1R protein (sCSF1R) was identified by plasma proteome profiling, and its role in evaluating LCH prognosis was explored. We prospectively measured plasma sCSF1R levels in 104 LCH patients and 10 healthy children using ELISA. Plasma sCSF1R levels were greater in LCH patients than in healthy controls (p < .001) and significantly differed among the three disease extents, with the highest level in MS RO+ LCH patients (p < .001). Accordingly, immunofluorescence showed the highest level of membrane-bound CSF1R in MS RO+ patients. Furthermore, the plasma sCSF1R concentration at diagnosis could efficiently predict the prognosis of LCH patients treated with standard first-line treatment (AUC = 0.782, p < .001). Notably, dynamic monitoring of sCSF1R levels could predict relapse early in patients receiving BRAF inhibitor treatment. In vitro drug sensitivity data showed that sCSF1R increased resistance to Ara-C in THP-1 cells expressing ectopic BRAF-V600E. Overall, the plasma sCSF1R level at diagnosis and during follow-up is of great clinical importance in pediatric LCH patients.


Subject(s)
Histiocytosis, Langerhans-Cell , Receptor, Macrophage Colony-Stimulating Factor , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/pathology , Histiocytosis, Langerhans-Cell/blood , Male , Female , Child , Prognosis , Child, Preschool , Infant , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Adolescent , Prospective Studies , Follow-Up Studies
2.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681709

ABSTRACT

Hepatitis B virus (HBV) infection is a major risk factor for hepatocellular carcinoma (HCC) development and is a global public health issue. High performance biomarkers can aid the early detection of HCC development in HBV-infected individuals. In addition, advances in the understanding of the pathogenesis of HBV infection and in clinical laboratory techniques have enabled the establishment of disease-specific tests, prediction of the progression of liver diseases, including HCC, and auxiliary diagnosis of HCC, using blood-based methods instead of biopsies of liver or HCC tissues. Viral factors such as the HBV genotype, HBV genetic mutations, HBV DNA, and HBV-related antigens, as well as host factors, such as tumor-associated proteins and post-translational modifications, especially glycosylated proteins, can be blood-based, disease-specific biomarkers for HCC development in HBV-infected patients. In this review, we describe the clinical applications of viral biomarkers, including the HBV genome and glycosylated proteins, for patients at a risk of HBV-related HCC, based on their molecular mechanisms. In addition, we introduce promising biomarker candidates for practical use, including colony stimulating factor 1 receptor (CSF1R), extracellular vesicles, and cell-free, circulating tumor DNA. The clinical use of such surrogate markers may lead to a better understanding of the risk of disease progression and early detection of HCC in HBV-infected patients, thereby improving their prognosis.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/pathology , Hepatitis B virus/genetics , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/etiology , Circulating Tumor DNA/blood , Extracellular Vesicles/metabolism , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/metabolism , Hepatitis B virus/isolation & purification , Hepatitis B virus/metabolism , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Humans , Liver Neoplasms/etiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood
3.
Eur J Appl Physiol ; 120(10): 2301-2309, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32767113

ABSTRACT

OBJECTIVE: Pulmonary rehabilitation is a cornerstone treatment in the management of chronic obstructive pulmonary disease (COPD). Acute bouts of exercise can lead to short bursts of inflammation in healthy individuals. However, it is unclear how COPD patients respond to acute bouts of exercise. This study assessed inflammatory responses to exercise in COPD patients at the start (phase 1) and end (phase 2) of pulmonary rehabilitation. METHODS: Blood samples were collected before and after an acute exercise bout at the start (phase 1, n = 40) and end (phase 2, n = 27) of pulmonary rehabilitation. The primary outcome was change in fibrinogen concentrations. Secondary outcomes were changes in CRP concentrations, total/differential leukocyte counts, markers of neutrophil activation (CD11b, CD62L and CD66b), and neutrophil subsets (mature, suppressive, immature, progenitor). RESULTS: Acute exercise (phase 1) did not induce significant changes in fibrinogen (p = 0.242) or CRP (p = 0.476). Total leukocyte count [mean difference (MD), 0.5 ± 1.1 (109 L-1); p = 0.004], neutrophil count [MD, 0.4 ± 0.8 (109 L-1); p < 0.001], and immature neutrophils (MD, 0.6 ± 0.8%; p < 0.001) increased post-exercise. Neutrophil activation markers, CD11b (p = 0.470), CD66b (p = 0.334), and CD62L (p = 0.352) were not significantly altered post-exercise. In comparison to the start of pulmonary rehabilitation (phase 2), acute exercise at the end of pulmonary rehabilitation led to a greater fibrinogen response (MD, 84 mg/dL (95% CI - 14, 182); p = 0.045). CONCLUSION: An acute bout of exercise does not appear to induce significant alterations in the concentrations of inflammatory mediators but can increase white blood cell subsets post-exercise. A greater fibrinogen response to acute exercise is seen at the end of pulmonary rehabilitation when compared to the start. Further research is required to understand the clinical context of these acute inflammatory responses to exercise.


Subject(s)
Exercise Therapy/methods , Pulmonary Disease, Chronic Obstructive/therapy , Aged , Biomarkers/blood , C-Reactive Protein/analysis , Female , Fibrinogen/analysis , Humans , L-Selectin/blood , Lung/physiopathology , Male , Middle Aged , Neutrophils/immunology , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/rehabilitation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood
4.
Clin Cancer Res ; 23(19): 5703-5710, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28655795

ABSTRACT

Purpose: Binding of colony-stimulating factor 1 (CSF1) ligand to the CSF1 receptor (CSF1R) regulates survival of tumor-associated macrophages, which generally promote an immunosuppressive tumor microenvironment. AMG 820 is an investigational, fully human CSF1R antibody that inhibits binding of the ligands CSF1 and IL34 and subsequent ligand-mediated receptor activation. This first-in-human phase I study evaluated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of AMG 820.Experimental Design: Adult patients with relapsed or refractory advanced solid tumors received intravenous AMG 820 0.5 mg/kg once weekly or 1.5 to 20 mg/kg every 2 weeks until disease progression, adverse event (AE), or consent withdrawal.Results: Twenty-five patients received ≥1 dose of AMG 820. AMG 820 was tolerated up to 20 mg/kg; the MTD was not reached. One dose-limiting toxicity was observed (20 mg/kg; nonreversible grade 3 deafness). Most patients (76%) had treatment-related AEs; the most common were periorbital edema (44%), increased aspartate aminotransferase (AST; 28%), fatigue (24%), nausea (16%), increased blood alkaline phosphatase (12%), and blurred vision (12%). No patients had serious or fatal treatment-related AEs; 28% had grade ≥3 treatment-related AEs. Grade 3 AST elevations resolved when treatment was withheld. AMG 820 showed linear pharmacokinetics, with minimal accumulation (<2-fold) after repeated dosing. Pharmacodynamic increases in serum CSF1 concentrations and reduced numbers of skin macrophages were observed. Best response was stable disease in 8 patients (32%).Conclusions: AMG 820 was tolerated with manageable toxicities up to 20 mg/kg every 2 weeks. Pharmacodynamic response was demonstrated, and limited antitumor activity was observed. Clin Cancer Res; 23(19); 5703-10. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Drug-Related Side Effects and Adverse Reactions/pathology , Neoplasms/drug therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/blood , Dose-Response Relationship, Drug , Female , Humans , Interleukin-1/blood , Macrophage Colony-Stimulating Factor/blood , Macrophage Colony-Stimulating Factor/genetics , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/genetics , Neoplasms/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Treatment Outcome
5.
Congenit Anom (Kyoto) ; 51(4): 183-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103458

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic cytokine that has neurotrophic and neuroprotective functions. However, its function in the mid- to late-gestational fetus remains unclear. We used enzyme-linked immunosorbent assay to analyze GM-CSF levels in cerebrospinal fluid (CSF), serum, and amniotic fluid of mouse fetuses. We also examined GM-CSF and receptor α (GM-CSFRα) levels in the fetal brain, liver, and placenta. GM-CSF peaked between embryonic day (E) 14 and E15 in the CSF. GM-CSF level was higher in the fetal serum than in the dam serum on E13 and decreased thereafter. GM-CSF and GM-CSFRα levels peaked between E13 and E15 in the brain. These results suggest that GM-CSF plays stage- and organ-specific roles in fetal development.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Amniotic Fluid/metabolism , Animals , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fetus/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/cerebrospinal fluid , Liver/metabolism , Mice , Mice, Inbred ICR , Organ Specificity , Placenta/metabolism , Pregnancy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood
7.
Gastroenterology ; 141(1): 208-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21557945

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) refers to 2 chronic inflammatory diseases of the intestine, ie, ulcerative colitis and Crohn's disease. IBD results from environmental factors (eg, bacterial antigens) triggering a dysregulated immune response in genetically predisposed hosts. Although the basis of IBD is incompletely understood, a number of recent studies have implicated defective innate immune responses in the pathogenesis of IBD. In this regard, there is much interest in therapies that activate innate immunity (eg, recombinant granulocyte-macrophage colony-stimulating factor). METHODS: In this study, we screened expression and function of circulating leukocyte granulocyte-macrophage colony-stimulating factor receptor (CD116) messenger RNA and surface protein in 52 IBD patients and 52 healthy controls. RESULTS: Our results show that both granulocyte and monocyte CD116 levels, but not CD114 or interleukin-3Rα, were significantly decreased in IBD compared to control (P<.001) and disease controls (irritable bowel syndrome; P<.001; rheumatoid arthritis; P<.025). IBD-associated CD116 repression was more prominent in patients with ulcerative colitis compared to Crohn's disease (P<.05), was independent of disease activity (P>.05), and was not influenced by current medications (P>.05). Receiver operating characteristic curve analysis revealed that leukocyte CD116 expression is a sensitive (85%) and specific (92%) biomarker for IBD. Moreover, granulocyte CD116-mediated function (phosphorylation of signal transducers and activators of transcription 3) paralleled decreased expression of CD116 in IBD granulocytes compared to control (P<.001). CONCLUSIONS: These studies identify defective expression and function of CD116 as a distinguishing feature of IBD and implicate an associated defect in innate immune responses toward granulocyte-macrophage colony-stimulating factor.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Immunity, Innate , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Adult , Biomarkers/blood , Case-Control Studies , Colitis, Ulcerative/blood , Colitis, Ulcerative/genetics , Colorado , Crohn Disease/blood , Crohn Disease/genetics , Down-Regulation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocytes/immunology , Humans , Immunity, Innate/genetics , Male , Middle Aged , Phosphorylation , Predictive Value of Tests , Prognosis , RNA, Messenger/blood , ROC Curve , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , STAT3 Transcription Factor/metabolism , Severity of Illness Index
8.
Ann Clin Lab Sci ; 38(4): 331-7, 2008.
Article in English | MEDLINE | ID: mdl-18988925

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophage-CSF (GM-CSF) are the principal cytokines in granulopoiesis and differentiation of granulocytic precursors. Their physiologic effects are mediated by binding to specific cell surface receptors (G-CSFr and GM-CSFr, respectively), which are widely expressed from immature bone marrow cells to mature peripheral granulocytes. The fact that concentrations of plasma G-CSF and GM-CSF and their receptors are changed in infectious diseases showing neutrophilia is known, but such changes in patients with chronic myelogenous leukemia (CML) have not been studied. Based on quantitative assays of plasma G-CSF and GM-CSF and their receptors on the peripheral granulocytes of CML patients and healthy controls, this study analyzes the differences between these groups in G-CSF and GM-CSF levels, as well as quantitative and qualitative changes in the receptors. Plasma levels of G-CSF and GM-CSF were measured in 47 patients in the chronic phase of CML and 25 healthy adults as normal controls. G-CSFr and GM-CSFr on peripheral granulocytes were analyzed by quantitative flow cytometry, and changes in G-CSF and GM-CSF receptor counts were also measured. Plasma concentrations of G-CSF and GM-CSF in CML patients were similar to normal controls (p>0.05). The quantity of G-CSFr on neutrophils was more highly expressed than on other cell types in both groups, and the amount of this receptor in patients with CML was less than in normal controls (p=0.001). GM-CSFr was expressed in higher concentrations on monocytes than neutrophils, and there was no difference in the amount of GM-CSFr on neutrophils. After incubation with excess G-CSF, the expressed amounts of G-CSFr on neutrophils and monocytes were decreased in both groups. However, G-CSFr on the monocytes was decreased in healthy controls (p=0.02) with no difference in patients with CML. The quantities of GM-CSFr expression on neutrophils and monocytes after incubation with excess GM-CSF were decreased in both groups. Granulocyte counts in peripheral blood of CML patients were not correlated with the plasma concentrations of G-CSF or GM-CSF, nor with the expression of G-CSFr or GM-CSFr on granulocytes. Granulopoiesis in patients with CML was not mediated by increased plasma CSF concentrations, and there was no difference in the amounts of G-CSFr or GM-CSFr expressed on the granulocytes. This suggests that a structural change may occur on monocytes of CML patients, since the binding capacity of G-CSFr to G-CSF on the monocytes is different from the normal controls.


Subject(s)
Granulocyte Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Receptors, Granulocyte Colony-Stimulating Factor/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Adult , Case-Control Studies , Female , Flow Cytometry , Granulocytes/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukocytes/metabolism , Male , Monocytes/metabolism , Neutrophils/metabolism
9.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 14(2): 225-7, 2006 Apr.
Article in Chinese | MEDLINE | ID: mdl-16638185

ABSTRACT

To evaluate soluble GM-CSF-Ralpha expression in patients with acute myeloid leukemia (AML) and its clinic significance, plasma concentration of solGM-Ralpha in de novo 66 patients with AML was detected by enzyme-linked immuno-sorbent assay, and the relationship between solGM-Ralpha levels and various clinical parameters was analyzed. The result showed that the levels of solGM-Ralpha in plasma of patients with AML were significantly higher than that in plasma of normal controls; the lowest level of solGM-Ralpha was found in plasma of patients with AML-M3 (3897.75 +/- 2651.43 pg/ml), the highest level of solGM-Ralpha was observed in plasma of patients with AML-M5 (9990.92 +/- 6325.43 pg/ml). Patients with high level of solGM-Ralpha were generally accompanied with a distinct clinical picture, including higher counts of white blood cell and myeloid precursors, as well as higher expression of CD34, CD95 and CD116 antigen. It is concluded that the high level of solGM-Ralpha in plasma of patients may suggest AML poor prognosis and play a role in pathogenesis of leukemia, the GM-CSF and its receptor solGM-Ralpha needs further study.


Subject(s)
Leukemia, Myeloid, Acute/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Adolescent , Adult , Aged , Antigens, CD34/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Prognosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , fas Receptor/blood
10.
Br J Haematol ; 121(1): 86-93, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12670335

ABSTRACT

The receptor for granulocyte-macrophage colony-stimulating factor (GM-CSF) can exist as both transmembrane (tmGMRalpha) and soluble (solGMRalpha) isoforms, and the latter, is a normal constituent of human plasma. We investigated if aberrant solGMRalpha expression occurs in haematopoietic malignancies and whether or not solGMRalpha expression levels correlated with clinical presentation. Compared with the normal population, patients with acute lymphoblastic leukaemia (ALL) had low levels of solGMRalpha whereas clonal disorders of the myeloid lineage demonstrated higher levels of solGMRalpha. Patients with acute myelogenous leukaemia (AML) and high levels of solGMRalpha presented with a distinct clinical picture. These patients were older, predominantly belonged to the M4 and M5 French-American-British (FAB) subtypes, and they had higher white blood cell counts at presentation including myeloid precursors and myeloblasts. They often presented with either unexplained lung infiltrates or hypoxia and lower rates of microbiologically defined infections. Elevated solGMRalpha levels were not associated with decreased relapse-free and overall survival in the AML population. On multivariate analysis, the correlation between elevated solGMRalpha levels and age, M4 and M5 FAB subtypes and decreased numbers of infections persisted. Our study is the first to describe that distinct clinical presentations are associated with aberrant solGMRalpha levels in haematological malignancies.


Subject(s)
Leukemia/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Case-Control Studies , Child , Disease-Free Survival , Female , Humans , Hypoxia/blood , Immunoglobulin Fab Fragments/blood , Leukemia, Myeloid, Acute/blood , Leukocyte Count , Lung Diseases/blood , Male , Middle Aged , Multivariate Analysis , Opportunistic Infections/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Survival Rate
11.
Exp Hematol ; 30(3): 187-94, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11882355

ABSTRACT

OBJECTIVE: The aim of this study was to determine whether granulocytes from patients with paroxysmal nocturnal hemoglobinuria (PNH) are more or less intrinsically sensitive to spontaneous apoptosis than granulocytes from healthy individuals. Resistance to apoptosis has been suggested as an explanation for the proliferation or selection of PNH clones. PATIENTS AND METHODS: Peripheral blood granulocytes from five patients with PNH, five patients with myelodysplastic syndrome (MDS), and five healthy volunteers were cultured in the absence of serum. Spontaneous apoptosis of the granulocytes was assessed every 6 hours by flow cytometry. The expression levels of CD16b, CD95, and granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor also were studied by flow cytometry, and caspase-3 activity was measured by fluorometry. RESULTS: There were no significant differences in the proportion or absolute numbers of apoptotic and apoptotic/dead granulocytes between the cells from PNH patients and healthy individuals, whereas those from MDS patients showed significantly lower frequencies of apoptotic granulocytes compared with normal controls. The proportion of CD16b(-) granulocytes was not significantly different among the three groups during in vitro culture. CD95 and GM-CSF receptor was not significantly increased in cultured granulocytes or noncultured granulocytes from, respectively, patients with PNH and normal controls. Caspase-3 activity significantly decreased in cultured granulocytes from MDS patients, but not in granulocytes from PNH patients. CONCLUSIONS: Granulocytes from PNH patients did not display a reduced sensitivity to spontaneous apoptosis, suggesting that the apoptosis of blood cells in PNH may not be an important factor in proliferation or selection of PNH clones. These findings are in agreement with the normal lifespan of granulocytes in vivo.


Subject(s)
Apoptosis , Granulocytes/pathology , Hemoglobinuria, Paroxysmal/blood , Adult , Aged , Annexin A5/blood , Antibodies, Monoclonal , CD59 Antigens/blood , Caspase 3 , Caspases/blood , Cells, Cultured , Culture Media, Serum-Free , Erythrocytes/immunology , Flow Cytometry , Fluorescent Antibody Technique , Granulocytes/enzymology , Granulocytes/immunology , Humans , Male , Middle Aged , Myelodysplastic Syndromes/blood , Propidium , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Receptors, IgG/blood , fas Receptor/blood
12.
Yonsei Med J ; 43(1): 43-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11854931

ABSTRACT

We analyzed the comparative amounts of granulocyte-colony stimulating factor (G-CSFr) and granulocyte macrophage CSF (GM-CSFr) receptors expressed on neutrophils and monocytes in measles patients to investigate the role of these CSFrs in the development of leukopenia including neutropenia and monocytopenia in measles. EDTA-anticoagulated peripheral blood of 19 measles patients, 10 children with other infections showing leukopenia and 16 children with normal complete blood cell counts (CBC)s were analyzed using flow cytometry and QuantiBRITE. The leukocyte (5260 +/- 2030/uL vs. 9900 + 2680/uL, p=0.000), neutrophil (2580 +/- 960/uL vs. 4250 +/- 2750/uL, p=0.024) and the lymphocyte counts of measles patients (1810 +/- 1430/uL vs. 4530 +/- 3450/uL, p= 0.006) were lower than in the normal controls. The neutrophils of measles patients expressed similar amounts of G- CSFr (1858 +/- 355) as normal children (1764 +/- 477, p= 0.564) and leukopenic patients (1773 +/- 673, p=0.713), but lower levels of GM-CSFr (535 +/- 118) than normal children (957 +/- 344, p=0.000) and leukopenic patients (832 +/- 294, p=0.002). The monocytes of measles patients expressed similar amounts of G-CSFr (916 +/- 336) and GM-CSFr (3718 +/- 906) as normal children (1013 +/- 391 and 4125 (2645, p > 0.05) but less than leukopenic patients (1454 +/- 398 and 5388 +/- 806, p > 0.05). The neutrophil and monocyte counts of measles patients did not correlate with the amount of G-CSFr or GM-CSFr expressed on neutrophils or monocytes (p > 0.05), but in the normal children, the monocyte count correlated with the levels of GM-CSFr on monocytes (r=0.951, p=0.049). In conclusion, neutropenia is one of the more important characteristics of measles patients, which could be due to the decreased GM-CSFr expression on neutrophils. However, the monocytopenia found in measles patients is not due to the decreased expression of CSFr on the monocytes.


Subject(s)
Measles/blood , Monocytes/chemistry , Neutrophils/chemistry , Receptors, Granulocyte Colony-Stimulating Factor/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Humans , Leukocyte Count , Neutropenia/etiology
13.
Yonsei Medical Journal ; : 43-47, 2002.
Article in English | WPRIM (Western Pacific) | ID: wpr-71380

ABSTRACT

We analyzed the comparative amounts of granulocyte-colony stimulating factor (G-CSFr) and granulocyte macrophage CSF (GM-CSFr) receptors expressed on neutrophils and monocytes in measles patients to investigate the role of these CSFrs in the development of leukopenia including neutropenia and monocytopenia in measles. EDTA-anticoagulated peripheral blood of 19 measles patients, 10 children with other infections showing leukopenia and 16 children with normal complete blood cell counts (CBC)s were analyzed using flow cytometry and QuantiBRITE. The leukocyte (5260 +/- 2030/uL vs. 9900 + 2680/uL, p=0.000), neutrophil (2580 +/- 960/uL vs. 4250 +/- 2750/uL, p=0.024) and the lymphocyte counts of measles patients (1810 +/- 1430/uL vs. 4530 +/- 3450/uL, p= 0.006) were lower than in the normal controls. The neutrophils of measles patients expressed similar amounts of G- CSFr (1858 +/- 355) as normal children (1764 +/- 477, p= 0.564) and leukopenic patients (1773 +/- 673, p=0.713), but lower levels of GM-CSFr (535 +/- 118) than normal children (957 +/- 344, p=0.000) and leukopenic patients (832 +/- 294, p=0.002). The monocytes of measles patients expressed similar amounts of G-CSFr (916 +/- 336) and GM-CSFr (3718 +/- 906) as normal children (1013 +/- 391 and 4125 (2645, p > 0.05) but less than leukopenic patients (1454 +/- 398 and 5388 +/- 806, p > 0.05). The neutrophil and monocyte counts of measles patients did not correlate with the amount of G-CSFr or GM-CSFr expressed on neutrophils or monocytes (p > 0.05), but in the normal children, the monocyte count correlated with the levels of GM-CSFr on monocytes (r=0.951, p=0.049). In conclusion, neutropenia is one of the more important characteristics of measles patients, which could be due to the decreased GM-CSFr expression on neutrophils. However, the monocytopenia found in measles patients is not due to the decreased expression of CSFr on the monocytes.


Subject(s)
Humans , Leukocyte Count , Measles/blood , Monocytes/chemistry , Neutropenia/etiology , Neutrophils/chemistry , Receptors, Granulocyte Colony-Stimulating Factor/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood
14.
Leukemia ; 11(10): 1700-10, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9324292

ABSTRACT

A quantitative analysis of expression levels of GM-CSF receptors was performed by flow cytometry in different disease categories, ie AML (n = 72), ALL (n = 18), and MDS (n = 12), as well as 12 healthy volunteers, using three different unconjugated GM-CSF/R monoclonal antibodies (McAbs) (HGM-CSFR (CD116), M5D12, 4B5F5), and appropriate standards. By using the reference HGM-CSFR McAb, in healthy subjects we found detectable levels of GM-CSF/R on blood monocytes (mean MESF (molecules of equivalent soluble fluorochrome)/cell: 36.1 x 10[3]), neutrophils (mean MESF/cell: 7.4 x 10[3]), bone marrow (BM) myelo-monocytic precursors (MESF range for the myeloid component, ie promyelocytes, myelocytes, metamyelocytes: 11.7-40.5 x 10[3], and for the monocytic lineage: 25.7-69.2 x 10[3]), and in two distinct subsets of BM CD34+ progenitor cells (GM-CSF/R dim: 2.5 x 10[3] MESF/cell, GM-CSF/R bright (10% of the total number of CD34 cells: 22.0 x 10[3] MESF/cell). In these subjects, there was no correlation between the expression levels of GM-CSF/R and CFU (CFU-GM, CFU-GEMM, BFU-E) colony production. Among the AML samples, M5D12 McAb was positive in 33%, 4B5F5 McAb in 90%, and HGM-CSF/R McAb in 78% of the cases examined (range of MESF/cell for the HGM-CSFR McAb: 0.9 x 10[3]-106.7 x 10[3]). The highest MESF values were seen in the M5 FAB subvariety (mean: 39.4 x 10[3]), where all the patients tested (n = 20) showed a strong positivity for the HGM-CSFR McAb. On the contrary, all ALL samples were GM-CSF/R negative except in two patients, who displayed a dim GM-CSF/R positivity (My+ALL: 1.3 x 10[3] MESF/cell; pro-B ALL: 1.0 x 10[3] MESF/cell). In most (>70%) M1 FAB subtypes, GM-CSF/R+ blasts co-expressed CD34low, HLA-DRhigh, CD33, CD38 antigens, and had little or no capacity to form CFU-GM colonies. GM-CSF/R+ blasts from the M5 FAB category were also positive for CD14, CD11c, CD33 and CD87. Furthermore, the number of GM-CSF/R expressed by leukemic cells from five out of 72 (7%) AML patients was above the highest values seen in normal samples (>69.2 x 10[3] MESF/cell), allowing the possibility of using this marker for the monitoring of the minimal residual disease (MRD) in a subset of AML. Cell culture studies aimed at evaluating GM-CSF receptor modulation following AML blast exposure to rhGM-CSF showed two distinct patterns of response; in the first group (6/10 cases) rhGM-CSF down-modulated GM-CSF receptors, whereas in the second group (4/10 cases), rhGM-CSF treatment was associated with either an increase or no change in the number of GM-CSF/R. In conclusion, cellular GM-CSF/R expression was variable and ranged from undetectable (ALL and a minority of AML) to very high intensities in M5 AML, and were also documented in some M0 AML, thus suggesting the concept that GM-CSF/R detection may be of help in lineage assignment of undifferentiated forms. Since the number of GM-CSF/R on AML blasts may be modulated after GM-CSF treatment, it can be postulated that the clinical use of GM-CSF in this disease may be optimized by a dynamic analysis of the number and the affinity status of GM-CSF-R in blasts and normal hemopoietic cells.


Subject(s)
Hematopoietic Stem Cells/ultrastructure , Leukemia, Myeloid/metabolism , Lymphocytes/ultrastructure , Myelodysplastic Syndromes/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Acute Disease , Adolescent , Adult , Flow Cytometry , Humans , Middle Aged , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood
15.
Stem Cells ; 15(4): 275-85, 1997.
Article in English | MEDLINE | ID: mdl-9253111

ABSTRACT

The detection of functional growth factor (GF) receptors on subpopulations of hemopoietic cells may provide a further dissection of immature cell subsets. Since little information is available about coexpression of different GF receptors at the level of single hemopoietic cells, we studied the feasibility of simultaneous cell staining with a combination of biotin- and digoxigenin-labeled GFs for flow cytometric detection of functional receptors. Using this methodology, coexpression of Kit and receptors for erythropoietin (EPO), interleukin 6 (IL-6), and GM-CSF on hemopoietic cells was studied by triple-staining of rhesus monkey bone marrow (BM) cells with labeled GFs and antibodies against other cell surface markers. Most of the immature, CD34+2 cells were Kit+ but did not display detectable levels of EPO-receptors (EPO-Rs) or GM-CSF-R. Approximately 60% of these CD34+2/Kit+ cells coexpressed the IL-6-R, demonstrating that immature cells are heterogeneous with respect to IL-6-R expression. Maturation of monomyeloid progenitors, as demonstrated by decreasing CD34 and increasing CD11b expression, is accompanied by a decline of Kit and an increase in GM-CSF-R expression in such a way that Kit+/GM-CSF-R+ cells are hardly detectable. IL-6-R expression is maintained or even increased during monomyeloid differentiation. IL-6-R and GM-CSF-R were not identified on most CD71+2 cells, which indicated that these receptors are probably not expressed during erythroid differentiation. Together with previous results, our data show that both Kit and CD71 are upregulated with erythroid commitment of immature progenitors. Upon further differentiation, Kit+/EPO-R-cells lose CD34 and acquire EPO-R. Maturing erythroid cells eventually lose CD71 and Kit expression but retain the EPO-R. In conclusion, this approach enables further characterization of the specificity of GFs for different bone marrow subpopulations. Apart from insight into the differentiation stages on which individual GFs may act, information about receptor coexpression may be used to identify individual cells that can respond to multiple GFs, and allows for further characterization of the regulation of lineage-specific differentiation.


Subject(s)
Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , Receptors, Erythropoietin/biosynthesis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, Interleukin/biosynthesis , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Antigens, Surface/metabolism , Bone Marrow/metabolism , Bone Marrow Cells , Cell Line , Digoxigenin , Humans , Immunophenotyping , Interleukin-6/metabolism , Macaca mulatta , Proto-Oncogene Proteins c-kit/blood , Receptors, Erythropoietin/blood , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Receptors, Interleukin/blood , Staining and Labeling , Stem Cell Factor/metabolism
17.
Leuk Lymphoma ; 24(1-2): 11-25, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9049958

ABSTRACT

The production of peripheral blood cells is regulated by hemopoietic growth factors (HGF) which promote the survival of stem cells and stimulate the proliferation and maturation of progenitors as well as effector functions of mature blood cell subsets. The actions of HGF's are determined by the cellular distribution of receptors for these HGF's within the hemopoietic tissues and by the functional program that receptor-expressing cells can execute after growth factor stimulation. Identification of stem cells and their progeny and delineation of the growth factor receptor phenotype of these cells will establish target cell range and functions of individual growth factors in hemopoiesis. Cells with specific HGF receptors can be detected and isolated by flow cytometric methods, e.g., by staining with biotinylated ligand and fluorescently-tagged streptavidin. Receptor-expressing cells can be classified on the basis of expression of the CD34 antigen and other markers that distinguish immature progenitors from more differentiated cells. Using this approach distinct expression patterns have been shown for the receptors for interleukin-3 (IL-3), IL-6, granulocyte/macrophage colony-stimulating factor (GM-CSF) and Steel Factor (SF) on subsets of CD34+ and CD34- cells in bone marrow. Expression of the IL-3 receptor (R), IL-6R and GM-CSFR appears to be very low on the most immature subsets of CD34+ cells, but increases progressively during successive stages, of in particular myelomonocytic differentiation. In contrast, the receptor for SF, i.e., Kit, is highly expressed on very immature CD34-bright/HLA-DR-dull cells, which include stem cells. Kit levels decline during myelomonocytic and B-lymphoid differentiation whereas they increase to maximal levels during early stages of erythropoiesis. The heterogeneity in receptor expression, together with other immunophenotypic characteristics, allows for the identification of distinct progenitor cell subsets and differentiation stages within the CD34+ cell compartment. By selecting appropriate phenotypic criteria it will be possible to further dissect the stem cell compartment and eventually establish the, possibly heterogeneous, HGF receptor phenotype of pluripotent stem cells.


Subject(s)
Antigens, CD34/blood , Hematopoietic Stem Cells/chemistry , Receptors, Colony-Stimulating Factor/blood , Animals , B-Lymphocyte Subsets/chemistry , B-Lymphocyte Subsets/immunology , Erythroid Precursor Cells/chemistry , Erythroid Precursor Cells/immunology , Hematopoietic Stem Cells/immunology , Humans , Leukemia, Myelomonocytic, Chronic/immunology , Leukemia, Myelomonocytic, Chronic/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/blood , Receptors, Interleukin/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...