Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Food Chem Toxicol ; 40(12): 1849-61, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12419700

ABSTRACT

Chloramphenicol (CAP) is haemotoxic in man, inducing two forms of toxicity. First, a commonly-occurring, dose-related, reversible bone marrow depression, which develops during treatment. Second, a rarer aplastic anaemia (AA), developing after treatment, is irreversible, and often fatal. Thiamphenicol (TAP) was developed as a replacement for CAP; however, there are no toxicological investigations in the mouse or rat on the dose-related haemotoxicity of TAP, in repeat dose gavage studies. Therefore, we have conducted a comprehensive investigation in these species, administering TAP for 7-17 days, to define haematological changes. Female BALB/c mice were gavaged with TAP, daily for 7-17 days at 400-1500 mg/kg; female Wistar Hanover rats were dosed with TAP daily at 50-375 mg/kg for 9 or 10 days. Haematological changes were studied at 1, 7 and 14 days post-dosing. In mice at day 1, TAP caused decreases in RBC, HCT and Hb; reticulocytes and platelets were reduced; changes were dose-related and reversible. Marrow cell counts were reduced; marrow was hypocellular, with erythroid depletion and progenitor cell vacuolation; the myeloid/erythroid (M:E) ratio was increased. In the rat, changes were not as clear-cut; there was anaemia with indications of reduced reticulocyte and platelet counts, and evidence of decreased neutrophils and lymphocytes. Marrow erythroid cells were decreased, precursor cells vacuolated, and the M:E ratio increased. We conclude that TAP induced haematological changes in the mouse and rat, parallelling the dose-dependent, reversible marrow depression reported in man; TAP is more haemotoxic in the rat than in the mouse.


Subject(s)
Anemia, Aplastic/chemically induced , Anti-Bacterial Agents/toxicity , Hematopoietic Stem Cells/drug effects , Thiamphenicol/toxicity , Anemia, Aplastic/pathology , Animals , Anti-Bacterial Agents/administration & dosage , Apoptosis/drug effects , Blood Cell Count , Blood Chemical Analysis , Dose-Response Relationship, Drug , Female , Hematocrit , Hemoglobins/drug effects , Mice , Mice, Inbred BALB C , Platelet Count , Random Allocation , Rats , Rats, Wistar , Reticulocytes/drug effects , Species Specificity , Thiamphenicol/administration & dosage
2.
Int J Hematol ; 76 Suppl 2: 163-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12430919

ABSTRACT

Aplastic anaemia seems to be predominantly a defect of the stem cell rather than the stroma, though abnormalities of the microenvironment may co-exist. There is highly suggestive evidence that the stem cell is the target of an immune attack, though the main evidence remains the response to immunosuppression with antilymphocyte globulin and cyclosporin. The stem cell defect remains even after recovery of the peripheral blood counts and the AA marrow is a fertile environment for the emergence of abnormal clones, particularly PNH. However, it has recently become apparent that there is an overlap with the myelodysplastic syndromes and clones of monosomy 7 and trisomy 8 amongst others are not uncommon in aplastic anaemia. Recent work has suggested that the emergence of a clone of monosomy 7 cells carries a poor prognosis, whereas trisomy 8 has a good prognosis particularly in response to cyclosporin. However, the setting in which monosomy 7 arises may affect the phenotypic expression. The immune targeting of stem cells may be associated with increased apoptosis in aplastic anaemia, in part mediated by fas expression, but not exclusively. Understanding the pathophysiology of AA should help to improve and perhaps target therapy.


Subject(s)
Anemia, Aplastic/pathology , Anemia, Aplastic/drug therapy , Anemia, Aplastic/etiology , Clone Cells/pathology , Hematopoietic Stem Cells/pathology , Humans , Immunosuppressive Agents/therapeutic use
3.
Clin Lab Haematol ; 24(6): 329-35, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12452812

ABSTRACT

In aplastic anaemia (AA), correction of bone marrow (BM) stromal function may contribute to the outcome of bone marrow transplantation (BMT). Engraftment of BM stromal cells is rarely observed, but engraftment of accessory cells (macrophages and T cells) may be important. We have improved a method of combined immunocytochemistry and FISH described by van Tol et al. (1998) to define the cellular origin and time course of engraftment of BM stromal accessory cells after sex-mismatched BMT. Long-term bone marrow cultures were trypsinized and cytospin preparations stained by immunocytochemistry using monoclonal antibodies against specific cell lineages followed by FISH for X and Y chromosomes. Low level phase contrast microscopy was used to study staining of individual cells simultaneously with fluorescence microscopy to define chromosomal pattern. In controls, the combined procedure did not affect the intensity of APAAP staining or the accuracy of sex chromosome determination. In cultures from AA patients after sex-mismatched BMT, cell lineages could be identified and donor or recipient origin determined unequivocally. This procedure enabled us to examine the origin (host/donor) of different cell lineages with high confidence, in addition to producing images of the combined staining.


Subject(s)
Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Stromal Cells/chemistry , Adult , Anemia, Aplastic/pathology , Anemia, Aplastic/therapy , Antigens, CD34 , Bone Marrow Transplantation , Cell Lineage , Chromosomes, Human, X , Chromosomes, Human, Y , Female , Graft Survival , Hematopoietic Stem Cells/pathology , Humans , Interphase , Male , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Middle Aged , Stromal Cells/ultrastructure , T-Lymphocyte Subsets/pathology , Transplantation Chimera , Transplantation, Homologous
4.
Int J Obstet Anesth ; 11(4): 301-5, 2002 Oct.
Article in English | MEDLINE | ID: mdl-15321534

ABSTRACT

A case of secondary pulmonary hypertension complicating twin pregnancy and necessitating caesarean section is presented. A planned team approach involving senior consultants in obstetrics, cardiology, neonatology and cardiac and obstetric anaesthesia resulted in successful perioperative management. Continuous spinal anaesthesia was the chosen technique to give adequate operating conditions with least haemodynamic disturbance. This is the first report of continuous spinal anaesthesia in such a condition.

5.
Br J Haematol ; 115(3): 642-52, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11736949

ABSTRACT

Defects in stromal cell function have been demonstrated in a number of aplastic anaemia (AA) patients. Here we have studied a patient with severe AA and abnormal stromal cell function who underwent bone marrow transplantation (BMT). The objective of this study was to investigate the timing and the mechanism of correction of the stromal defect after transplantation. The patient, a 25-year-old woman with severe AA, underwent BMT from her brother. BM was obtained from the patient on five occasions: 2 weeks pre BMT, and 3, 8, 16 and 21 months post BMT. Stromal cells were grown to confluence and recharged with purified CD34+ cells from normal donors. The support of such cells, as assessed by weekly colony-forming assay (CFU) of non-adherent cells, was compared with that of stromal layers grown from normal BM. A novel technique of combined fluorescence in situ hybridization (FISH) and immunocytochemistry was used to determine the origin of specific stromal cell types on cytospins of stroma post BMT. Stromal function was defective at 2 weeks pre BMT and at 3 months post BMT, but returned to normal at 8 and 16 months post BMT. At 21 months post BMT, stromal fibroblasts and endothelial cells were shown to be of recipient origin, and macrophages and T cells were of donor origin. We present here evidence in a case of severe AA for defective stromal function before BMT and delayed normalization of function after BMT. This correlated with engraftment of donor macrophages and T cells, but not fibroblasts and endothelial cells.


Subject(s)
Anemia, Aplastic/therapy , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Pregnancy Complications, Hematologic/therapy , Adult , Anemia, Aplastic/immunology , Colony-Forming Units Assay , Endothelium/immunology , Female , Fibroblasts/immunology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Macrophages/immunology , Pregnancy , Pregnancy Complications, Hematologic/immunology , T-Lymphocytes/immunology , Time Factors , Transplantation, Homologous
6.
Br J Haematol ; 115(1): 167-74, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11722430

ABSTRACT

The changes in bone marrow (BM) stem cell reserve and function and stromal cell function in patients with active systemic lupus erythematosus (SLE) were investigated. The study was carried out on seven SLE patients and 28 healthy controls using flow cytometry and in vitro cell culture assays. We found that patients had low CD34(+) cells, compared with the control group, reflecting the decrease of both CD34(+)/CD38(-) and CD34(+)/CD38(+) cells. Patient CD34(+)/Fas(+) but not CD34(-)/Fas(+) cells were significantly increased. Apoptotic (7AAD(dim)) cells were higher among CD34(+)/Fas(+) than among CD34(+)/Fas(-) cells, and individual values of apoptotic CD34+ cells strongly correlated with the number of CD34(+)/Fas(+) cells. These findings are suggestive of a Fas-mediated apoptosis accounting for the low CD34(+) cells in SLE patients. Moreover, we found that patients had low numbers of granulocyte-macrophage colony-forming units (CFU-GM) and erythroid burst-forming units (BFU-E), compared with the control group, and that the generation of colony-forming cells in long-term BM cultures was significantly reduced. Patient BM stroma failed to support allogeneic progenitor cell growth. In one patient, CD34(+) cells were increased, apoptotic CD34(+)/Fas(+) cells were normalized and defective stromal cell function was restored after autologous stem cell transplantation. We concluded that defective haemopoiesis in SLE patients is probably caused, at least in part, to the presence of autoreactive lymphocytes in BM.


Subject(s)
Antigens, CD34/analysis , Bone Marrow Cells/immunology , Lupus Erythematosus, Systemic/immunology , Stem Cells/immunology , Adult , Analysis of Variance , Apoptosis , Case-Control Studies , Cells, Cultured , Colony-Forming Units Assay , Female , Flow Cytometry , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans , Immunohistochemistry , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/surgery , Male , Middle Aged , Statistics, Nonparametric , Time Factors , fas Receptor/analysis
7.
Br J Haematol ; 113(3): 706-12, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380462

ABSTRACT

Aplastic anaemia (AA) is a syndrome of haemopoietic failure involving increased apoptosis in stem cells. AA CD34+ cells often have upregulated Fas antigen, but this does not explain the increased apoptosis in all patients. To examine whether abnormal expression of the apoptotic modulators Bcl-2 and Bcl-x is involved in increased apoptosis in the CD34+ cells of patients, we examined cells from 19 AA patients and 18 normal controls by triple staining for CD34, Bcl-2 or Bcl-x, together with 7-amino actinomycin D to determine viability or with staining for Fas antigen. We confirmed increased apoptosis of CD34+ cells in patients. All CD34+ cells in patients and controls expressed Bcl-2 and Bcl-x with no significant difference between the groups. In patients, viability of CD34+/Bcl-2hi cells was similar to that of CD34+/Bcl-2lo cells, but CD34+/Bcl-xhi cells were significantly more viable than CD34+/Bcl-xlo cells. CD34+ cells from AA patients expressed upregulated Fas antigen, but this did not correlate with Bcl-2 or Bcl-x expression. These results suggest a more significant role for Bcl-x as an anti-apoptotic regulator in CD34+ cells in AA than Bcl-2. The induction of death by Fas antigen may bypass the anti-apoptotic effect of Bcl-2 and Bcl-x in CD34+ cells in AA.


Subject(s)
Anemia, Aplastic/metabolism , Antigens, CD34 , Bone Marrow Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , fas Receptor/metabolism , Adolescent , Adult , Aged , Anemia, Aplastic/immunology , Apoptosis , Case-Control Studies , Child , Female , Flow Cytometry , Humans , Immunophenotyping , Male , Middle Aged , bcl-X Protein
8.
Eur J Haematol ; 67(4): 245-51, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11860446

ABSTRACT

OBJECTIVE: To investigate further the cellular defect responsible for impaired granulopoiesis in severe congenital neutropenia (SCN), we have evaluated bone marrow (BM) stem cell reserve and function and BM stromal cell myelopoiesis supporting capacity in two patients with SCN. METHODS: BM primitive stem cells and myeloid progenitor cells were assessed using flow cytometry, limiting dilution assay, clonogenic assays, and long-term BM cultures (LTBMC). BM stroma function was assessed by evaluating the ability of irradiated stromal layers from the patients to induce granulocyte-macrophage colony formation (CFU-GM) by normal CD34+ cells. RESULTS: Compared to the normal controls (n = 37), SCN patients displayed a low percentage of CD34+/CD38+ cells (P < 0.05), low CFU-GM colony formation by highly purified CD34+ cells (P < 0.05), low CFU-GM recovery in LTBMC (P < 0.05), and normal primitive stem cells as indicated by the frequency of CD34+/CD38- cells and the number of long-term culture initiating cells. Patient BM stromal layers exhibited normal myelopoiesis supporting capacity as shown by the CFU-GM content of irradiated LTBMC recharged with normal CD34+ cells. In addition, patient LTBMC supernatants displayed 20-fold normal granulocyte colony stimulating factor and 2-fold normal granulocyte-macrophage colony stimulating factor levels. CONCLUSION: These data show that primitive BM stem cells and stromal cells are not affected in SCN patients, while they support further the concept of a primary defect at the myeloid progenitor cell level. To know the differentiation stage at which the underlying defect causes the malfunction will be relevant for further elucidation of its nature at the molecular level.


Subject(s)
Bone Marrow/pathology , Cytokines/metabolism , Hematopoietic Stem Cells/pathology , Neutropenia/congenital , Stromal Cells/pathology , Adult , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured/drug effects , Coculture Techniques , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Female , Flow Cytometry , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Humans , Male , Neutropenia/pathology , Stromal Cells/metabolism
9.
Stem Cells ; 18(6): 428-34, 2000.
Article in English | MEDLINE | ID: mdl-11072031

ABSTRACT

It has previously been shown that patients with aplastic anemia (AA) have a stem cell defect both of proliferation and differentiation. This has been shown by long-term bone marrow (BM) culture, long-term initiating cell assays, and committed progenitor assays. We present, for the first time, data on megakaryocyte (Mk) colony formation from purified BM CD34(+) cells from patients with AA. The results are compared with those from normal controls and from patients with paroxysmal nocturnal hemoglobinuria (PNH) and the myelodysplastic syndromes (MDSs). Those treated for AA had previously received immunosuppression (antithymocyte globulin and/or cyclosporin). No patients had received bone marrow transplantation. A total of 13 AA patients (five untreated, eight treated), six PNH, six MDS, and 13 normal donors were studied. BM CD34(+) cells were purified by indirect labeling and then cultured in a collagen-based Mk assay kit (MegaCult-C, StemCell Technologies). The cultures were fixed on day 12, and the Mk colonies were identified by the alkaline phosphatase anti-alkaline phosphatase technique using the monoclonal antibody CD41 (GP IIb/IIIa). The slides were scored for Mk colony-forming units (CFU-Mks) (3-20 and >20 cells), Mk burst-forming units (BFU-Mks) (>50 cells), and mixed colonies. The results show that total Mk colony formation in AA was significantly lower than in normal donors (p<0.0001), both in untreated patients/nonresponders to treatment (p = 0.0001) and in complete/partial responders (p<0.002). There was no significant difference in Mk colony formation in treated and untreated patients (p = 0.05). Patients with AA had a lower total colony formation than PNH patients (p = 0.0002). PNH patients exhibited lower colony formation than normal controls (p = 0.03), as shown by MDS patients, although the considerable number of variables resulted in a lack of statistically significant difference from normal controls (p = 0.2). We have now shown that Mk colony formation from purified BM CD34(+) cells is significantly reduced, supporting previous evidence that AA results from a stem cell defect.


Subject(s)
Anemia, Aplastic/blood , Hematopoietic Stem Cells/cytology , Hemoglobinuria, Paroxysmal/blood , Megakaryocytes/cytology , Myelodysplastic Syndromes/blood , Cell Differentiation , Cell Division , Cell Separation , Humans
10.
Blood ; 96(9): 3272-5, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11050013

ABSTRACT

To investigate whether bone marrow (BM) stem cell compartment and/or BM microenvironment are affected by the immune insult in autoimmune cytopenias (AICs), BM stem cell reserve and function and BM stromal function were studied in 15 AIC patients. Stem cells were evaluated by means of flow cytometry, clonogenic progenitor cell assays, long-term BM cultures (LTBMCs), and limiting dilution assay for quantification of long-term-culture initiating cells (LTC-ICs). Stromal cell function was assessed with the use of preformed irradiated LTBMCs from patients and normal controls, recharged with normal CD34(+) cells. AIC patients exhibited a high number of CD34(+), CD34(+)/CD38(+), and CD34(+)/CD38(-) cells; high frequency of granulocyte-macrophage colony forming units in the BM mononuclear cell fraction; high colony recovery in LTBMCs; and normal LTC-IC frequency. Patient BM stromal layers displayed normal hematopoietic-supporting capacity and increased production of granulocyte-colony stimulating factor. Data from this study support the concept that AIC patients with severe, resistant disease might be appropriate candidates for autologous stem cell transplantation.


Subject(s)
Autoimmune Diseases/pathology , Bone Marrow Cells/pathology , Hematologic Diseases/pathology , Hematopoietic Stem Cells/pathology , Stromal Cells/pathology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adult , Antigens, CD/analysis , Antigens, CD34/analysis , Antigens, Differentiation/analysis , Autoimmune Diseases/immunology , Bone Marrow Cells/immunology , Cells, Cultured , Colony-Forming Units Assay , Female , Hematologic Diseases/immunology , Hematopoietic Stem Cells/immunology , Humans , Male , Membrane Glycoproteins , Middle Aged , NAD+ Nucleosidase/analysis , Stromal Cells/immunology
11.
Anesth Analg ; 91(1): 58-61, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10866887

ABSTRACT

UNLABELLED: We compared the hemodynamic effects of a bolus administration of 1 microg/kg remifentanil for 1, 3, and 5 min (1, 0.33, and 0.2 microg. kg(-1). min(-1), respectively) in patients scheduled for coronary artery bypass grafting anesthetized with small-dose propofol. The study was terminated after only eight patients had been enrolled (three received remifentanil at a rate of 1.0 microg. kg(-1). min(-1), two at 0.33 microg. kg(-1). min(-1), and three at 0.2 microg. kg(-1). min(-1)) because of severe hemodynamic instability, which was particularly marked in four patients and consisted of severe bradycardia in one patient and severe hypotension with a reduction in systemic vascular resistance in three others. One patient showed evidence of myocardial ischemia. All patients responded to therapeutic interventions. The results show that remifentanil should be given only by slow infusion to such patients. IMPLICATIONS: This study investigates the effect on the heart and blood vessels of various rates of administration of boluses of a relatively new potent opiate, remifentanil, to patients with coronary artery disease. The results show that remifentanil should be given only by slow infusion to such patients.


Subject(s)
Anesthetics, Intravenous/adverse effects , Hemodynamics/drug effects , Piperidines/adverse effects , Anesthetics, Intravenous/administration & dosage , Bradycardia/chemically induced , Coronary Artery Bypass , Depression, Chemical , Humans , Hypotension/chemically induced , Piperidines/administration & dosage , Remifentanil
12.
Bone ; 27(1): 29-40, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10865206

ABSTRACT

Osteoclast formation from hemopoietic precursors is induced by TRANCE (also called RANKL, ODF, and OPGL), a membrane-bound ligand expressed by bone marrow stromal cells. Because soluble recombinant TRANCE is a suboptimal osteoclastogenic stimulus, and to eliminate the need for such dependence on stromal cells, membrane-bound TRANCE was expressed in hematopoietic precursors using retroviral gene transfer. Four TRANCE-expressing osteoclast cell lines were established that continuously generate large numbers of multinucleated cells and express tartrate-resistant acid phosphatase and calcitonin receptors. The multinuclear cells are long-lived and either fuse continuously with each other and with mononuclear cells to form enormous syncytia, or separate to form daughter multinuclear cells. When formed on bone, but not on plastic, the majority of multinuclear cells develop actin rings on bone, and resorb bone, suggesting that bone matrix may provide additional signals that facilitate osteoclastic functional maturation. Surprisingly, multinuclear cells originate from fusion of proliferating mononuclear cells that strongly express the mature macrophage markers F4/80 and Fc receptor, which are not expressed by osteoclasts. These results indicate that osteoclasts can be derived from F4/80-positive and Fc receptor-positive cells, and that TRANCE induces osteoclastic differentiation partly by suppressing the macrophage phenotype.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Carrier Proteins/physiology , Cell Lineage/physiology , Membrane Glycoproteins/physiology , Osteoclasts/cytology , Animals , Cell Differentiation/physiology , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Humans , Male , Mice , Osteoclasts/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Retroviridae
13.
Br J Haematol ; 108(3): 582-91, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10759717

ABSTRACT

The mechanism of action of antithymocyte globulin (ATG) in the treatment of aplastic anaemia (AA) and myelodysplastic syndromes (MDS) is poorly understood and may involve many different mechanisms. The aim of this in vitro study was to investigate further the effect of ATG on haemopoietic progenitor cells. A total of 16 patients (10 AA and 6 MDS) and 12 normal control subjects were studied. Purified bone marrow (BM) CD34+ cells were cultured in committed progenitor assay in the presence of ATG and autologous serum, then scored on day 14 for granulocyte-monocyte colony-forming units (CFU-GM) and erythroid colonies. ATG was found to be inhibitory to haemopoietic progenitor cells at high concentrations (1000 microg/ml and 100 microg/ml). This was confirmed by CD34-FITC and 7AAD staining of purified normal CD34+ cells after overnight incubation with ATG. In contrast, at lower doses (0.1-10 microg/ml), ATG produced an increase in colony growth in most normal, MDS and AA BM CD34+ cells. The greatest effect was in patients with non-severe AA, in whom the greatest increase in CFU-GM was seen at 0.5 microg/ml (P < 0.02) and 0.1 microg/ml (P = 0.02) and erythroid colonies at 0.1 microg/ml (P < 0.05). Serum ATG levels peaked during infusion to levels that were found to be toxic to haemopoietic progenitor cells in vitro and fell thereafter to levels that were associated with the highest colony numbers (0.1 and 0.5 microg/ml) in vitro. These results suggest that an increase in haemopoietic progenitor cells by ATG may be one of several important mechanisms for haematological recovery in AA and MDS.


Subject(s)
Anemia, Aplastic/therapy , Antigens, CD34/immunology , Antilymphocyte Serum/therapeutic use , Hematopoietic Stem Cells/drug effects , Myelodysplastic Syndromes/therapy , Adolescent , Adult , Aged , Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Case-Control Studies , Cell Division/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology
14.
Br J Haematol ; 111(4): 1164-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11167757

ABSTRACT

The mechanism of action of anti-thymocyte globulin (ATG) in aplastic anaemia (AA) is complex. Bone marrow (BM) CD34(+) cells in AA have been shown to be more apoptotic and have a higher expression of Fas antigen (Fas-ag) than in normal donors. The aims of this study were to delineate further the mechanism for increased bone marrow progenitor cell apoptosis in AA and investigate the effects of ATG on apoptosis and Fas-ag expression. BM was obtained from six normal donors and 10 untreated AA patients. We confirmed that AA BM CD34(+) cells were more apoptotic than normal donor cells (P = 0.002). Following treatment with ATG, the mean percentage reduction of apoptosis was 34% (9.2-65.9%). BM from 30 AA and 10 normal donors was then stained for CD34, Fas-ag and 7-AminoActinomycin D. The proportion of CD34(+) Fas(+) cells was higher in untreated AA (P = 0.0001) than in normal donors. Results also showed that the majority of CD34(+) Fas(+) cells were apoptotic/dead in normal donors (mean 81%) and AA (88%), indicating that Fas is involved in apoptosis of CD34(+) cells. In contrast, the majority of CD34(+) Fas(-) cells in normal donors were live (mean 91%), while two patterns emerged in untreated AA. In seven patients, the majority of cells were live, however, in the remaining eight patients, the majority of cells were apoptotic/dead, suggesting an alternative mechanism for apoptosis in addition to Fas-ag. Finally, we have shown that in vivo ATG treatment reduced the expression of Fas-ag on AA BM CD34(+) cells.


Subject(s)
Anemia, Aplastic/therapy , Antigens, CD34 , Apoptosis/physiology , Hematopoietic Stem Cells/physiology , Immunoglobulins, Intravenous/therapeutic use , Adolescent , Adult , Aged , Anemia, Aplastic/blood , Case-Control Studies , Fas Ligand Protein , Female , Fluorescent Antibody Technique , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Male , Membrane Glycoproteins/metabolism , Middle Aged , Statistics, Nonparametric , fas Receptor/metabolism
15.
Br J Haematol ; 107(4): 797-803, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606887

ABSTRACT

Transforming growth factor beta (TGF-beta) 1 is a ubiquitous bifunctional cytokine implicated in the regulation of haemopoietic stem cells and bone marrow stromal cells. We analysed sera from 63 patients with aplastic anaemia and describe a significant reduction of TGF-beta1 that was directly related to their treatment status. Untreated patients (n = 35), patients who did not respond (n = 15) and those with a partial response (n = 23) to treatment had significantly lower TGF-beta1 than the normal control group (n = 55), P < 0.0001, P < 0.0001 and P = 0.002 respectively. Patients in complete remission (n = 15) exhibited TGF-beta1 serum levels comparable to the control group. In addition, there was a correlation (r = 0.83, P < 0.0001) between serum TGF-beta1 and platelet count at time of sample. We have demonstrated that the primary source of TGF-beta1 in peripheral blood mononuclear cell (PBMC) cultures was not CD3-positive cells. These data indicate aplastic anaemia is associated with a decreased TGF-beta1 expression in peripheral blood circulation, which may be a direct consequence of thrombocytopenia. In vitro stromal layers grown from aplastic patient bone marrow (n = 14) produced significantly lower levels of TGF-beta1 (P = 0.02) when compared to normal stroma (n = 15). In the aplastic anaemia bone marrow compartment we postulate that accessory cells down-regulate TGF-beta1 expression to allow stem cell cycling to counteract hypoplasia. As TGF-beta1 is important in the regulation of haemopoiesis, dysregulation of this cytokine in combination with previously described abnormal cytokine expression may contribute significantly to the pathophysiology of aplastic anaemia by exacerbating primary stem cell defects.


Subject(s)
Anemia, Aplastic/blood , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Aged , Anemia, Aplastic/therapy , Cells, Cultured , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Platelet Count , Stromal Cells/metabolism
16.
Blood ; 91(10): 3582-92, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9572992

ABSTRACT

Improved survival in aplastic anemia (AA) has shown a high incidence of late clonal marrow disorders. To investigate whether accelerated senescence of hematopoietic stem cells might underlie the pathophysiology of myelodysplasia (MDS) or paroxysmal nocturnal hemoglobinuria (PNH) occurring as a late complication of AA, we studied mean telomere length (TRF) in peripheral blood leukocytes from 79 patients with AA, Fanconi anemia, or PNH in comparison with normal controls. TRF lengths in the patient group were significantly shorter for age than normals (P < .0001). Telomere shortening was apparent in both granulocyte and mononuclear cell fractions, suggesting loss at the level of the hematopoietic stem cell. In patients with acquired AA with persistent cytopenias (n = 40), there was significant correlation between telomere loss and disease duration (r = -.685; P < .0001), equivalent to progressive telomere erosion at 216 bp/yr, in addition to the normal age-related loss. In patients who had achieved normal full blood counts (n = 20), the rate of telomere loss had apparently stabilised. There was no apparent association between telomere loss and secondary PNH (n = 13). However, of the 5 patients in the study with TRF less than 5.0 kb, 3 had acquired cytogenetic abnormalities, suggesting that telomere erosion may be relevant to the pathogenesis of MDS in aplastic anemia.


Subject(s)
Anemia, Aplastic/genetics , Telomere/ultrastructure , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Anemia, Aplastic/blood , Blood Cell Count , Bone Marrow/pathology , Cell Division , Child , Disease Progression , Fanconi Anemia/blood , Fanconi Anemia/genetics , Female , Hematopoiesis , Hematopoietic Stem Cells/pathology , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/etiology , Hemoglobinuria, Paroxysmal/genetics , Humans , Leukocytes/ultrastructure , Male , Middle Aged , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/genetics , Polymorphism, Restriction Fragment Length , Prognosis , Risk
17.
J Infect Dis ; 176(6): 1510-6, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9395362

ABSTRACT

Hematologic abnormalities are often seen in patients infected with human immunodeficiency virus (HIV). The effect of HIV infection of bone marrow stroma on support of uninfected CD34 progenitor cells in long-term bone marrow culture (LTBMC) was investigated. Results show that HIV-infected bone marrow stroma was unable to adequately support CD34 progenitor cells in vitro. Zidovudine or didanosine was added to cultures in an attempt to reverse the suppressive effects exerted by HIV and to determine whether such suppression was mediated by transfer of HIV infection to progenitor cells. Didanosine failed to reduce the suppressive effects of HIV, whereas zidovudine compounded the observed suppression. HIV infection of bone marrow stroma, while reducing the production of nonadherent cells, did not increase apoptosis and cell death in such cells. In contrast, zidovudine enhanced apoptosis and cell death in nonadherent cells produced by both HIV-infected and control LTBMC.


Subject(s)
Anti-HIV Agents/pharmacology , Bone Marrow Cells/virology , HIV/physiology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Antigens, CD34/analysis , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Death/drug effects , Cells, Cultured , Didanosine/pharmacology , HIV Core Protein p24/analysis , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/virology , Zidovudine/pharmacology
18.
Br J Haematol ; 97(1): 146-52, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9136957

ABSTRACT

The mechanism of release of CD34+ cells into the peripheral blood (PB) after mobilization treatment with chemotherapy and/or growth factors is not clearly understood. Growth factors may induce increased proliferation and self renewal within the stem cell compartment. It is possible that they alter adhesion molecule profiles or other progenitor:stroma interactions, to allow release of these cells into the periphery. However, CD34+ cells are present in the PB under steady-state conditions, albeit in low number. Growth factors such as granulocyte colony-stimulating factor (G-CSF) may promote the survival of CD34+ cells in the PB by suppressing apoptosis. In order to test this hypothesis, we have quantitated apoptotic cells in the CD34+ fraction of peripheral blood stem cell (PBSC) collections, using two-colour flow cytometry, after staining with anti-CD34 antibody and the fluorescent DNA binding agent, 7-amino actinomycin D (7AAD). 7AAD differentially stains live, apoptotic and dead cells, due to the altered accessibility of DNA in each subpopulation. We have shown a significant reduction in the proportion of apoptotic cells in the CD34+ population mobilized by G-CSF compared to CD34+ cells in unstimulated PB, consistent with the theory that G-CSF is acting, at least in part, by suppressing apoptosis. In addition, we found that G-CSF mobilized CD34+ cells are less apoptotic than CD34+ cells of unstimulated normal bone marrow, indicating that, at the doses used, G-CSF is significantly altering the survival capacity of the mobilized cells.


Subject(s)
Antigens, CD34/metabolism , Apoptosis/physiology , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cells/cytology , Adolescent , Adult , Bone Marrow Cells , Female , Humans , Male , Middle Aged
19.
Br J Haematol ; 95(4): 605-10, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8982033

ABSTRACT

Endogenous serum thrombopoietin (TPO) levels were measured in 31 patients with aplastic anaemia (AA) using an enzyme immunoassay with a sensitivity of 20 pg/ ml. The median platelet count for all AA patients was 30 +/- 29 x 10(9)/l (range 5-102) compared with a median of 284 +/- 59 x 10(9)/l (range 148-538) for normal controls. Serum TPO levels were significantly elevated in all patients compared with normals (1706 +/- 1114.2, range 375-5000 v 78 +/- 54, range 16.5-312.9, P < 0.0001). There was no correlation between serum TPO levels and the degree of thrombocytopenia in AA patients, but TPO levels were significantly higher in patients who were platelet transfusion dependent than in patients who were transfusion independent (P < 0.01). There was a trend for higher TPO levels in patients with severe AA compared with non-severe AA patients. Clinical trials of TPO and a related truncated, pegylated molecule, megakaryocyte growth and development factor (PEG-rHuMGDF), are awaited to determine whether treatment with these drugs will result in increased platelet counts in patients with AA.


Subject(s)
Anemia, Aplastic/blood , Thrombopoietin/blood , Adult , Aged , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Platelet Count
20.
Br J Haematol ; 93(1): 30-7, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8611471

ABSTRACT

Haematological abnormalities are often seen in patients infected with HIV. A number of mechanisms are thought to contribute to this bone marrow suppression, including impaired stromal function and direct infection of progenitor cells. Evidence suggests that both bone marrow progenitor cells and perhaps stromal cells are open to infection by HIV, which raises the possibility that bone marrow stromal cells may serve as a reservoir for HIV. This study investigated the cellular targets and kinetics of in vitro infection of stroma in long-term bone marrow culture (LTBMC) using both mono- and lymphocytotropic strains of HIV-1. p24 ELISA and reverse transcriptase (RT) assay demonstrated that stroma could be infected with HIV and release infectious virions. The target cells for infection were shown to be macrophages by immunohistochemistry (APAAP), dual immunofluorescence staining (using CD68 and p24) and electron microscopy. The data show that it was possible to infect stroma in LTBMC with HIV and that such infection was productive. The main target for infection was bone marrow macrophages. In contrast to peripheral blood derived macrophages, these cells were susceptible to both lymphocytotropic and monocytotropic strains of HIV-1. The data suggests that these bone marrow macrophages may act as a reservoir for HIV, Infection of bone marrow macrophages may affect haemopoiesis either by transmission of HIV infection to developing progenitor cells through direct cell-to-cell contact or by altering the ability of the stroma to support normal development.


Subject(s)
Bone Marrow/virology , HIV-1/pathogenicity , Macrophages/virology , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microscopy, Electron , Stromal Cells/virology , Time Factors , Tropism
SELECTION OF CITATIONS
SEARCH DETAIL
...