Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Blood ; 95(1): 342-6, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10607722

ABSTRACT

To assess the relative contribution of genetic factors in the variation of F cells (FC) and other hematologic variables, we conducted a classical twin study in unselected twins. The sample included 264 identical (monozygotic [MZ]) twin pairs and 511 nonidentical (dizygotic [DZ]) same-sex twin pairs (aged 20 to 80 years) from the St. Thomas' UK Adult Twin Register. The FC values were distributed continuously and positively skewed, with values ranging from 0.6% to 22%. FC values were higher in women than in men and decreased with age, with the variables accounting for 2% of the total FC variance. The intraclass correlations of FC values were higher in MZ (rMZ = 0.89) than in DZ (rDZ = 0.49) twins. The XmnI-(G)gamma polymorphism in the beta-globin gene cluster had a significant effect on FC levels, accounting for approximately 13% of the total FC variance. Variance components analysis showed that the FC values were accounted for predominantly by additive genetic and nonshared environmental influences, with an estimate of heritability of 0.89. Hemoglobin levels and red blood cell, white blood cell, and platelet numbers were also substantially heritable, with heritability estimates of 0.37, 0.42, 0.62, and 0.57, respectively. Previously, studies of sib pairs with sickle cell disease and isolated family studies showed that high levels of Hb F and FC tend to be inherited. Here, our classical twin study demonstrated that the variance of FC levels in healthy adults is largely genetically determined. (Blood. 2000;95:342-346)


Subject(s)
Erythrocytes/classification , Globins/genetics , Twins, Dizygotic/genetics , Twins, Monozygotic/genetics , Adult , Aged , Cohort Studies , Erythrocyte Count , Female , Genotype , Hemoglobins/metabolism , Humans , Leukocyte Count , Male , Middle Aged , Models, Genetic , Phenotype , Platelet Count , Polymorphism, Genetic , Registries , Sex Factors , United Kingdom
3.
Am J Hum Genet ; 62(6): 1468-74, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9585587

ABSTRACT

Fetal hemoglobin (Hb F) and fetal cell (FC) levels in adults show considerable variation and are influenced by several genetic variants; the major determinants appear to be unlinked to the beta-globin gene cluster. Recently, a trans-acting locus controlling Hb F and FC production has been mapped to chromosome 6q23 in an Asian Indian kindred that includes individuals with heterocellular hereditary persistence of Hb F (HPFH) associated with beta thalassemia. We have extended the kindred by 57 members, bringing the total studied to 210, and have saturated the region with 26 additional markers. Linkage analysis showed tight linkage of the quantitative-trait locus (QTL) to the anonymous markers D6S976 (LOD score 11.3; recombination fraction .00) and D6S270 (LOD score 7.4; recombination fraction .00). Key recombination events now place this QTL within a 1-2-cM interval spanning approximately 1.5 Mb between D6S270 and D6S1626. Furthermore, haplotype analysis has led to a reevaluation of the genealogy and to the identification of additional relationships in the kindred.


Subject(s)
Chromosomes, Human, Pair 6 , Fetal Hemoglobin/genetics , Quantitative Trait, Heritable , Chromosome Mapping , Female , Fetal Hemoglobin/biosynthesis , Genetic Linkage , Genotype , Haplotypes , Humans , Male , Microsatellite Repeats , Pedigree
4.
Leuk Lymphoma ; 22(1-2): 83-90, follow. 186, color plate VI, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8724532

ABSTRACT

There is now good evidence that tumour necrosis factor [TNF] stimulates DNA synthesis of B-chronic lymphocytic leukaemia (B-CLL) cells. The malignant clone produces TNF, and addition of exogenous TNF up-regulates the TNF mRNA in B-CLL cells. Interleukin-6 (rIL-6) may also be important in this growth loop. We studied the interaction of TNF and IL-6 in the regulation of DNA synthesis (3H-TdR uptake), cytokine release and cell survival in CLL cells in vitro. Addition of TNF (100 U/ml over 5 days) enhanced DNA synthesis from 718 +/- 284 (mean cpm +/- SE) to 2730 +/- 545 compared to cells cultured in medium alone (n = 16, p < 0.01). TNF-alpha induced DNA synthesis was inhibited in all cases studied by the addition of anti-TNF monoclonal antibody (5 micrograms/ml) to cell cultures. Spontaneous IL-6 protein release was enhanced in the presence of TNF (100 U/ml and 250 U/ml) by CLL cells at 48 hours of culture 143.6% and 172% (p < 0.05, n = 6). At 120 hours of culture, the increase was 323% and 412.5% (4 of 7 cases) of the control respectively. IL-6 (100 U/ml or greater) increased spontaneous DNA synthesis (3H-TdR uptake) but, in the presence of high concentrations of TNF-alpha, inhibited TNF induced DNA synthesis in a dose dependent manner. Cell survival was reduced in the presence of anti-IL-6 mAb, while IL-6 was able to protect CLL cells from spontaneous apoptosis. These results suggest that IL-6 in an autocrine manner may inhibit DNA synthesis but prolongs survival in CLL cells. Increased serum IL-6 levels were detected in 27 of 50 cases of CLL, the mean level being significantly higher in Rai Stage III and IV cases compared to Rai Stage O-II cases.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Growth Inhibitors/pharmacology , Interleukin-6/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplastic Stem Cells/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Division/drug effects , DNA Fragmentation , DNA Replication , DNA, Neoplasm/biosynthesis , Female , Humans , Immunophenotyping , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Interleukin-6/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
5.
Br J Haematol ; 91(4): 963-70, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8547149

ABSTRACT

Induction of tissue factor (TF) expression on monocytes and endothelial cells is central to the development of septic coagulopathy. Serum concentrations of endotoxin in septic patients who develop disseminated intravascular coagulation (DIC) do not, however, reach the levels that would directly stimulate TF expression on either monocytes or endothelium. We show, using an in vitro coculture system, that the interaction of monocytes with endothelium induces the expression of significant levels of TF. Unstimulated cocultures of monocytes (2 x 10(4)/well) and endothelial cells (2 x 10(4)/well) produced 35.3 +/- 8.5 mU of PCA/well, representing a 5-fold increase over the combined PCA of each cell type cultured alone (7.1 +/- 1.5 mU, n = 6, P < 0.001). Significant enhancement was also found in the presence of low concentrations of LPS. Induction of TF protein was confirmed by Western blotting. Fixation of monocytes with paraformaldehyde completely abolished TF induction in cocultures, whereas fixation of endothelium had no effect, suggesting that TF induction occurred in monocytes rather than endothelial cells. Induction of TF in cocultures could be further augmented by preincubating the endothelial cells with IFN-gamma. When endothelium was prestimulated with 500 U/ml IFN-gamma there was 142 +/- 11% increase over unstimulated cocultures (n = 5, P < 0.01). TF induction was inhibited by 32 +/- 6% in the presence of anti-ICAM-1 mAb (n = 5, P < 0.01). Our results suggest that monocyte interactions with vascular endothelium, regulated by inflammatory cytokines, and mediated by adhesive ligand binding, leads to the induction of functional monocyte TF protein, which may be responsible for the initiation of DIC in sepsis.


Subject(s)
Disseminated Intravascular Coagulation/etiology , Endothelium, Vascular/physiology , Monocytes/physiology , Thromboplastin/biosynthesis , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Adhesion/physiology , Cell Communication/physiology , Coculture Techniques , Disseminated Intravascular Coagulation/metabolism , Endothelium, Vascular/drug effects , Humans , Intercellular Adhesion Molecule-1/immunology , Interferon-gamma/pharmacology , Monocytes/metabolism
6.
Leuk Res ; 18(1): 55-60, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8289467

ABSTRACT

Previous studies have shown that interleukin-4 (IL-4) may have both stimulatory and inhibitory effects on the growth of normal and malignant B-cells in vitro. We studied the effects of IL-4 on tumour necrosis factor (TNF) induced and spontaneous proliferation (3H-TdR incorporation) and spontaneous release of TNF and interleukin-6 (IL-6) by purified B-cell chronic lymphocytic leukaemia (CLL) cells in vitro. TNF (100 U/ml) increased 3H-TdR uptake in cells to 700 +/- 302% of control (mean +/- S.E., n = 9, p = 0.033). Recombinant IL-4 (10 ng/ml) consistently inhibited DNA synthesis in all CLL patients studied. When added at the start of 5 day cultures, IL-4 inhibited both spontaneous (41 +/- 17% inhibition, n = 3) and TNF induced (46 +/- 5% inhibition, n = 9, p = 0.01) 3H-TdR uptake. Similar results were obtained when IL-4 was added after 48 h of culture. This effect of IL-4 was dose dependent. Inhibition was not related to clinical stage. IL-4 (whether added at T0 or T48h) also inhibited spontaneous release of TNF and IL-6 measured at 48 and 120 h. TNF and IL-4 had no consistent effect on normal cord blood CD5+ B-cells. These data show that IL-4 has inhibitory effects on B-CLL DNA synthesis and also inhibits spontaneous release of IL-6 and TNF in vitro. IL-4 may have a role in vivo in reducing proliferation in these B-cell malignancies by inhibiting potential autocrine growth loops.


Subject(s)
Cytokines/metabolism , Interleukin-4/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Division , Humans , Interleukin-6/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
Br J Haematol ; 83(3): 491-7, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8387324

ABSTRACT

Hepatitis C virus (HCV) is responsible for the majority of cases of post transfusion non-A non-B (NANB) hepatitis in thalassaemia major (TM). Twelve multi-transfused TM patients with serological, biochemical, histological and molecular biological evidence of HCV infection have been treated for 6 months with recombinant alpha-interferon (IFN). Ten (83%) responded as assessed by a fall of at least 50% of pre-treatment serum transaminase levels. Histological improvement was observed in 6/7 responders tested. Natural killer (NK) cell activity 24 h after the first dose of IFN was significantly increased in responders as compared to non-responders (P < 0.05). HCV RNA disappeared from serum in 5/12 and from liver tissue in 2/5 of the responders. The degree of induction of peripheral blood mononuclear cell 2'5' oligoadenylate synthetase messenger RNA (2-5 OAS mRNA), an enzyme induced by IFN, after the first dose of IFN did not correlate with response. IFN was generally well tolerated. We conclude that the response rate in multi-transfused TM patients infected with HCV and treated with IFN is similar to that in non-multi-transfused patients.


Subject(s)
Hepatitis C/therapy , Interferon-alpha/therapeutic use , beta-Thalassemia/complications , Adolescent , Adult , Base Sequence , Child , Chronic Disease , Female , Hepacivirus/genetics , Hepatitis C/blood , Hepatitis C/pathology , Humans , Interferon alpha-2 , Interferon-alpha/adverse effects , Killer Cells, Natural/pathology , Liver/pathology , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Viral/analysis , Recombinant Proteins , Transaminases/blood , Transfusion Reaction
9.
Br J Haematol ; 81(2): 231-4, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1379467

ABSTRACT

Tumour necrosis factor (TNF) can act as an autocrine growth factor for hairy cell leukaemia (HCL) cells. The TNF produced by the malignant clone may also inhibit normal haematopoiesis thereby contributing to the cytopenias observed in patients with the disease. We have studied the effects of infusing a murine monoclonal anti-TNF antibody in three patients with HCL. In two patients receiving 0.5 mg of antibody/kg on alternate days for 12 d, the drug was well tolerated. The third patient received 2 mg/kg on alternate days and developed symptoms of serum sickness by day 9. In two patients with severe B-lymphocytopenia, circulating CD19 and CD20 positive, B-cells were restored to normal, the majority of which were negative for the HCL-associated marker CD11c. B-lymphocyte recovery was associated with a rise in serum immunoreactive IL-6 and with an early rise in immunoreactive TNF. These short courses of anti-TNF MAb treatment had modest effect on the tumour burden, producing a reduction in splenomegaly in one patient. Exploration of the effects of more prolonged administration of higher dose anti-TNF antibody will only be feasible when less immunogenic MAbs are available.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Leukemia, Hairy Cell/therapy , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, CD/analysis , Antigens, CD19 , Antigens, CD20 , Antigens, Differentiation, B-Lymphocyte/analysis , B-Lymphocytes/immunology , CD11 Antigens , Humans , Immunoradiometric Assay , Interleukin-6/analysis , Leukemia, Hairy Cell/blood , Male , Mice , Middle Aged , Tumor Necrosis Factor-alpha/analysis
10.
Blood ; 79(11): 3059-66, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1316791

ABSTRACT

Cytomegalovirus (CMV) remains the most common single infective cause of death following allogeneic bone marrow transplantation (BMT) from major histocompatibility complex (MHC)-identical siblings, whereas Epstein-Barr virus (EBV)-related disease is infrequent. We show here that MHC-unrestricted cytotoxic effector cells in the peripheral blood of BMT recipients are highly effective at killing EBV-infected target cells, but are inactive against CMV-infected target cells. Differential cytotoxicity is associated with disparate target structure expression. Although both EBV- and CMV-infected target cells express viral antigens, it is only those infected with EBV that express the adhesion molecule lymphocyte function-associated antigen 1 (LFA1; CD11a/18). Thus, EBV-infected target cells are able to interact with the principal LFA1 ligand, intercellular adhesion molecule 1 (ICAM1; CD54), which is expressed on posttransplant peripheral blood mononuclear (PBM) effector cells. CMV-infected target cells cannot utilize this ligand. Posttransplant cytotoxicity against EBV-infected target cells is abolished by target and effector cell blockade with monoclonal antibodies (MoAbs) to LFA1 and ICAM1, respectively, demonstrating the functional relevance of this additional ligand interaction. These results provide an illustration both of the importance and of the limitations of MHC-unrestricted cytotoxicity in vivo and may explain the frequency of CMV disease and the relative rarity of EBV-related disease following allogeneic transplantation from MHC-matched siblings. The increased immunosuppression used following MHC-mismatched/matched unrelated-donor BMT may cause this MHC-unrestricted defense mechanism to fail and may contribute to the greatly increased incidence of EBV lymphoproliferative syndrome in these patients.


Subject(s)
Bone Marrow Transplantation , Cytomegalovirus/immunology , Cytotoxicity, Immunologic , HLA Antigens/immunology , Herpesvirus 4, Human/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Adolescent , Adult , Antibodies, Monoclonal , Bone Marrow Transplantation/adverse effects , Child , Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/immunology , Humans , Immune Tolerance , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Tumor Virus Infections/etiology , Tumor Virus Infections/immunology
11.
Blood ; 77(9): 1996-2001, 1991 May 01.
Article in English | MEDLINE | ID: mdl-1708296

ABSTRACT

Major histocompatibility complex-unrestricted lymphokine-activated killer (LAK) cells have been proposed as therapy for a variety of hematologic malignancies. Because these cells recognize and kill their targets independently of their antigen specific CD3 receptor, it is unclear how they might discriminate between normal and malignant cells. We now propose one such mechanism for the selective killing of myeloid leukemia blasts. While both CD2+ and CD2- activated killer cells may inhibit the clonogenic growth of myeloid leukemia cells, only the CD2+ subset effectively inhibits the growth of normal myeloid (granulocyte-macrophage and granulocyte) progenitors. This difference appears to reflect differential requirements for cell adhesion molecule recognition between normal and malignant progenitor cells. Inhibition of the growth of normal granulocyte-macrophage colonies by CD2+ LAK cells is blocked by antibodies to the CD2-lymphocyte function-associated antigen 3 (LFA-3) (CD58) cell adhesion system. In contrast, these antibodies have no effect on CD2+ LAK-mediated inhibition of malignant cell clonogenic growth. Instead, antibodies to the LFA-1 (CD11a/CD18)-intercellular adhesion molecule 1 (ICAM-1) (CD54) adhesion system reduce inhibition. These differences correspond to differential expression of the CD54 cell adhesion molecule by normal and malignant myeloid progenitor cells because less than 15% of normal CD34 positive cells are CD54+ while greater than 85% of CD34+ acute myeloid leukemia blasts express the CD54 antigen. LFA-3, the ligand for CD2, is strongly expressed by erythrocytes, and these cells competitively inhibit killing of normal but not malignant clonogenic cells in an analogous way to the effects of monoclonal antibody to the CD2-LFA-3 adhesion system. The operation of this effect in vivo may be a basis for selective cytotoxicity by CD2+ LAK against clonogenic myeloid blast cells, and could be exploited further with infusion of appropriate monoclonal antibodies.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Lymphokine-Activated/immunology , Leukemia, Myeloid, Acute/immunology , Antigens, CD/analysis , Antigens, CD/immunology , Antigens, CD34 , Antigens, Differentiation/analysis , Antigens, Differentiation/immunology , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Surface/immunology , CD2 Antigens , CD58 Antigens , Cell Adhesion/immunology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/immunology , Humans , Intercellular Adhesion Molecule-1 , Membrane Glycoproteins/immunology , Receptors, Immunologic/analysis , Receptors, Immunologic/immunology
12.
Blood ; 77(6): 1283-9, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-1900445

ABSTRACT

Cytokine-secreting, major histocompatibility complex-unrestricted activated killer (AK) cells are toxic to a wide range of virus-infected or malignant target cells and may be generated endogenously, eg, after bone marrow transplantation, or by infusion of cytokines such as recombinant interleukin-2 (rIL-2). Although AK cells secrete cytokines such as gamma-interferon and tumor necrosis factor, which are themselves able to recruit fresh cytokine-secreting AK cells, activation in both settings is short-lived, implying the existence of homeostatic regulatory mechanisms. We now demonstrate one mechanism by which rapid homeostasis is achieved. We show that IL-4 is produced in patients with both endogenously and exogenously generated AK cells. The cytokine was detected in serum after marrow transplantation, and IL-4 transcripts appeared in circulating lymphocytes during rIL-2 infusion. Although IL-4 inhibited the induction phase of AK cell function, it had no significant inhibitory effect on the ability of AK cells from these individuals to respond to restimulation. Nonetheless, neutralization of the IL-4 induced during cell activation doubled the half-life of AK function, once activating stimuli were removed, from 18 to 44 hours and produced a 2-log increase in AK cell secretion of tumor necrosis factor and gamma-interferon. These data suggest that IL-4 induced in vivo during lymphocyte activation abbreviates AK cell responses once the triggering stimuli have been removed. Neutralization of endogenous IL-4 in vivo by appropriate monoclonal antibodies might prolong the duration of AK function.


Subject(s)
Homeostasis/physiology , Interleukin-2/pharmacology , Interleukin-4/pharmacology , Killer Cells, Natural/physiology , Bone Marrow Transplantation , Homeostasis/drug effects , Humans , Infusions, Intravenous , Interferon-gamma/metabolism , Interleukin-2/administration & dosage , Interleukin-4/metabolism , Interleukin-4/physiology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/physiology , Lymphocytes/drug effects , Lymphocytes/physiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism
13.
Br J Haematol ; 77(2): 237-44, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2004026

ABSTRACT

Administration of interleukin 2 (IL2) to patients with minimal residual malignant disease following myeloablative chemo-radiotherapy may augment immune reconstitution and reduce the risk of relapse by increasing cytotoxic effector function and cytokine dependent killing directed at residual malignant cells. The ability of IL2 generated activated killer cells to inhibit haemopoietic progenitor cells and to release gamma-interferon (gamma IFN) and tumour necrosis factor (TNF) may, however, retard haemopoietic recovery, as both TNF and gamma IFN inhibit normal myelopoiesis in vitro. To determine the effect of IL2 infusion on myeloid regeneration in vivo, we have examined haemopoietic recovery in patients receiving this cytokine following autologous marrow transplantation or ablative chemotherapy. We find that IL2 infusion accelerates neutrophil recovery and that granulocyte-macrophage colony stimulating factor (GMCSF) and IL3 mRNA become detectable in circulating mononuclear cells. Induction of TNF by IL2 may also contribute to subsequent acceleration of myelopoiesis by initiation of GM-CSF mRNA synthesis in patient marrow fibroblasts. These results show that IL2 infusion may facilitate myeloid recovery when administered during the period of haemopoietic regeneration following ablative chemoradiotherapy.


Subject(s)
Bone Marrow/pathology , Hematopoietic Cell Growth Factors/blood , Interleukin-2/therapeutic use , Leukemia, Myeloid/therapy , Multiple Myeloma/therapy , Acute Disease , Adolescent , Adult , Aged , Blood Cell Count , Bone Marrow Transplantation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Interleukin-3/biosynthesis , Leukemia, Myeloid/blood , Male , Middle Aged , Multiple Myeloma/blood
15.
Br J Haematol ; 72(2): 122-6, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2503022

ABSTRACT

Culture of lymphocytes from allograft recipients or from normal donors with Interleukin 2 (IL2) induces high levels of gamma-interferon (gamma IFN) and tumour necrosis factor (TNF) secretion. We now show that production of these two cytokines can also be increased after autologous bone marrow transplantation (BMT) and induced following chemotherapy for haematological malignancy. These effects occur even though the regenerating IL2 responsive lymphocytes in this context have previously been extensively exposed to cytotoxic agents. IL2 induced secretion of gamma-IFN and TNF is higher in autograft recipients than in patients treated by chemotherapy alone. This effect may be related to differences in the phenotypic profile between recovering lymphocytes in the two groups and to an increased degree of prior in vivo lymphocyte activation in the autologous bone marrow transplant recipients. In both groups of patients, IL2 acts on CD3+ T cells and on CD16+ NK cells so that depletion of either subset incompletely abrogates IL2 dependent gamma-IFN secretion. As both TNF and gamma-IFN possess anti-leukaemic and anti-infective activity, enhancement of their secretion by IL2 infusion after autologous bone marrow transplant and induction after chemotherapy may be of therapeutic benefit.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation , Interleukin-2/pharmacology , Lymphocytes/drug effects , Adolescent , Adult , Aged , Cells, Cultured , Child , Hematologic Diseases/blood , Hematologic Diseases/therapy , Humans , Interferon-gamma/metabolism , Middle Aged , Tumor Necrosis Factor-alpha/metabolism
16.
Blood ; 73(5): 1351-8, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2495037

ABSTRACT

After marrow transplantation, major histocompatibility complex (MHC)-unrestricted natural killer (NK) lymphocytes are among the first cells to appear in the circulation. After T-cell-depleted bone marrow transplantation (TD-BMT), these cells have an activated pattern of target cell killing; they also secrete lymphokines including gamma-interferon (gamma-IFN), interleukin-2 (IL-2), and tumor necrosis factor (TNF) and may have a significant role as a primary defense against viral reactivation and in the elimination of residual host malignancy. We studied 43 patients with hematologic malignancy, treated by allogeneic TD-BMT, autologous nondepleted BMT, or chemotherapy alone to investigate (a) the mechanisms underlying the generation of these activated killer cells, (b) the range of conditions under which they are produced, and (c) their surface phenotype. We showed that gamma-IFN-secreting activated killer cells with the capacity to kill MHC-nonidentical NK-resistant targets are generated 4 to 6 weeks after either allogeneic TD-BMT or autologous BMT but do not appear after treatment with chemotherapy. Production therefore is not owing to T-cell depletion per se or to host donor alloreactivity, nor is it caused by stimulation by alloantigens contained in blood product support since no significant difference exists between allograft and chemotherapy patients in the number of units of blood platelet support given in the posttreatment period. Because most patients had no evidence of stimulation from virus reactivation/infection, the phenomenon of activation therefore appears to represent posttransplant immune disregulation following repopulation of the host immune system with lymphoid subsets derived exclusively from blood and marrow. Activated killing is predominantly mediated by the CD16+ CD3- subset, but substantial activity remains in the CD16- CD3+ cell fraction. Monoclonal antibodies (MoAbs) that block interaction with class-I MHC molecules at the level of target cell (W6/32 anti-HLA class I) or effector cell (CD8) do not inhibit killing by CD16- CD3+ cells. Activated killer cells may contribute to the lower risk of relapse after marrow transplantation as compared with intensive chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Leukemia/surgery , Lymphocyte Activation , Adolescent , Adult , Animals , Blood Transfusion , Child , Cricetinae , Histocompatibility Antigens Class I/genetics , Humans , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , Leukemia/drug therapy , Leukemia/therapy , Lymphocyte Depletion , Middle Aged , Phenotype , Transplantation, Autologous , Transplantation, Homologous , Virus Diseases/etiology
17.
Transplantation ; 45(6): 1084-91, 1988 Jun.
Article in English | MEDLINE | ID: mdl-2968009

ABSTRACT

Low-density cells (LDC) prepared from peripheral blood by fractionation over hypertonic metrizamide contain 95% of cells with veiled morphology, almost all of which are HLA-DR-positive and have characteristics of antigen-presenting cells. In normal individuals the monoclonal antibodies RFD1 and RFD2 divide these cells into three phenotypically distinct populations, D1+D2-, D1-D2+ and D1-D2-. The RFD1-positive population is nonphagocytic. We have investigated the recovery of LDC in peripheral blood after (T cell-depleted) marrow transplantation, to assess whether defects in antigen-presenting cell (APC) subpopulations could contribute to the prolonged immune-paresis of marrow graft recipients. We find that APC of donor origin and with apparently normal morphology, phenotype, and function appear within 6 weeks of BMT. By three months the donor-derived nonphagocytic RFD1-positive subset has disappeared, although phagocytic RFD2-positive cells remain. The disappearance of the RFD1-positive subset is associated with a loss of antigen presentation by patients' LDC of the soluble protein antigen tetanus toxoid, though the capacity to present alloantigen and stimulate in a mixed lymphocyte reaction is retained. Donor-derived RFD1-positive cells and soluble antigen-presenting capacity do not reappear for one year or more. This biphasic recovery of RFD1-positive cells contrasted with the continued production of RFD2-positive APC, implies that the phenotypic and functional distinction between APC subpopulations in peripheral blood also reflects a separate ontogeny. Since these marrow graft recipients retain the phagocytic (RFD2-positive) APC but lose the nonphagocytic (RFD1-positive) APC subset, there is now an opportunity to explore the role of each subset in antigen processing and presentation.


Subject(s)
Antigen-Presenting Cells/classification , Bone Marrow Transplantation , Phenotype , Adolescent , Adult , Antibodies, Monoclonal , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cell Count , Child , Female , HLA-DR Antigens/immunology , Humans , Longitudinal Studies , Lymphocyte Culture Test, Mixed , Male , Tetanus Toxoid/immunology , Tissue Donors , Transplantation, Homologous
18.
Lancet ; 1(8592): 969-71, 1988 Apr 30.
Article in English | MEDLINE | ID: mdl-2896830

ABSTRACT

Recombinant tumour necrosis factor (TNF) promotes survival and induces proliferation in the tumour cells from two malignancies of B lymphocytes--hairy-cell leukaemia and B-chronic lymphocytic leukaemia. Culture with TNF also induces TNF mRNA and protein, so the cytokine may act as an autocrine tumour growth factor. These growth promoting effects are antagonised by alpha but not by gamma interferon.


Subject(s)
Leukemia, Hairy Cell/pathology , Leukemia, Lymphoid/pathology , Tumor Necrosis Factor-alpha/pharmacology , B-Lymphocytes , Cell Division/drug effects , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Leukemia, Hairy Cell/metabolism , Leukemia, Lymphoid/metabolism , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor , Thymidine/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
19.
Br J Haematol ; 67(3): 273-9, 1987 Nov.
Article in English | MEDLINE | ID: mdl-3318910

ABSTRACT

After T-cell depleted marrow transplantation, there is a rapid recovery of cytotoxic effector cells, with activity against targets not susceptible to killing by 'resting' natural killer cells. These targets include Epstein-Barr virus transformed B cells and leukaemic cell lines. Activated killer cell function declines by 3 months after transplantation. We find that when CD3 negative effector cells are obtained from these patients and cultured in vitro with interleukin 2 there is a further enhancement of cytotoxic activity against a range of target cells in the early post-transplant period, and a restoration of high level cytotoxic activity to effector cells obtained 3 months or more after the procedure. These results may have relevance to attempts to reduce the incidence of leukaemic relapse, and EBV + ve lymphoma outgrowth after T-cell depleted BMT.


Subject(s)
Bone Marrow Transplantation , Interleukin-2/pharmacology , Killer Cells, Natural/physiology , T-Lymphocytes/immunology , Adolescent , Adult , Cell Separation , Child , Cytotoxicity Tests, Immunologic , Humans , Leukocytes, Mononuclear/immunology
20.
Eur J Immunol ; 17(1): 43-7, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3545853

ABSTRACT

Following T cell-depleted bone marrow transplantation, helper T cell numbers remain depressed for some months. Nonetheless, functional B cells can be adoptively transferred to the recipients of such grafts, where they continue to secrete antibody. We now show that immunoglobulin production by these transferred B cells is induced by activated large granular lymphocytes (LGL) which circulate in the recipients in substantial numbers during the immediate post-transplant period. The LGL are CD3 negative and therefore provide help in an antigen-unlinked manner. Helper effects for autologous (donor) B cells are augmented by the addition of anti-LFA-2 (anti-CD2) which appears to act by blocking recruitment of LGL inhibitory to developing B cells. In contrast antibody to the beta chain of LFA-1, which effectively reduces natural killer activity of LGL, does not influence their helper function. The peripheral blood LGL fraction thus contains both helper and cytotoxic activity, which can be distinguished by appropriate monoclonal antibodies.


Subject(s)
Antibody Formation , Bone Marrow Transplantation , Killer Cells, Natural/physiology , Lymphocyte Cooperation , Antigens, Surface/immunology , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , Female , Humans , Immunization, Passive , Lymphocyte Depletion , Male , Postoperative Period , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL