Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 52
1.
Neth Heart J ; 27(9): 414-425, 2019 Sep.
Article En | MEDLINE | ID: mdl-31111459

BACKGROUND: Machine learning (ML) allows the exploration and progressive improvement of very complex high-dimensional data patterns that can be utilised to optimise specific classification and prediction tasks, outperforming traditional statistical approaches. An enormous acceleration of ready-to-use tools and artificial intelligence (AI) applications, shaped by the emergence, refinement, and application of powerful ML algorithms in several areas of knowledge, is ongoing. Although such progress has begun to permeate the medical sciences and clinical medicine, implementation in cardiovascular medicine and research is still in its infancy. OBJECTIVES: To lay out the theoretical framework, purpose, and structure of a novel AI consortium. METHODS: We have established a new Dutch research consortium, the CVON-AI, supported by the Netherlands Heart Foundation, to catalyse and facilitate the development and utilisation of AI solutions for existing and emerging cardiovascular research initiatives and to raise AI awareness in the cardiovascular research community. CVON-AI will connect to previously established CVON consortia and apply a cloud-based AI platform to supplement their planned traditional data-analysis approach. RESULTS: A pilot experiment on the CVON-AI cloud was conducted using cardiac magnetic resonance data. It demonstrated the feasibility of the platform and documented excellent correlation between AI-generated ventricular function estimates as compared to expert manual annotations. The resulting AI solution was then integrated in a web application. CONCLUSION: CVON-AI is a new consortium meant to facilitate the implementation and raise awareness of AI in cardiovascular research in the Netherlands. CVON-AI will create an accessible cloud-based platform for cardiovascular researchers, demonstrate the clinical applicability of AI, optimise the analytical methodology of other ongoing CVON consortia, and promote AI awareness through education and training.

2.
Clin Radiol ; 71(12): 1313.e1-1313.e4, 2016 Dec.
Article En | MEDLINE | ID: mdl-27720180

AIM: To investigate the feasibility of using a low-concentration test bolus in abdominal aorta computed tomography (CT) angiography (CTA). MATERIALS AND METHODS: In 10 patients referred for CTA of the abdominal aorta with a body mass index (BMI) ≤28 kg/m2, a standard test bolus of 10 ml contrast medium (CM; 350 mg iodine/ml) was compared with a low-concentration test bolus (5 ml CM; 350 mg iodine/ml; 1:1 diluted with saline) in terms of time to peak enhancement (tPE) and peak enhancement (PE). RESULTS: No significant differences were found between the standard and low-concentration test bolus in terms of tPE and PE. CONCLUSIONS: A low-concentration test bolus (5 ml, 1:1 diluted with saline) is feasible in patients with a BMI ≤28 kg/m2.


Aorta, Abdominal/diagnostic imaging , Computed Tomography Angiography/methods , Contrast Media/administration & dosage , Multidetector Computed Tomography/methods , Radiographic Image Enhancement/methods , Triiodobenzoic Acids/administration & dosage , Feasibility Studies , Humans , Reproducibility of Results
3.
Clin Radiol ; 71(6): 615.e7-615.e13, 2016 Jun.
Article En | MEDLINE | ID: mdl-27059387

AIM: To evaluate image quality after contrast medium (CM) and tube voltage reduction in computed tomography angiography (CTA) of the pulmonary artery. MATERIALS AND METHODS: Thirty-three patients referred for CTA of the pulmonary artery for suspected pulmonary embolism were included. Patients were randomly assigned to Protocol I (100 ml of 350 mg iodine/ml iodinated CM; n=16) or Protocol II (50 ml of 350 mg iodine/ml iodinated CM; n=17). Dual-energy CT (80 kV and 140 kV) was performed in all patients. An averaged weighted series equivalent to a 120 kV image acquisition was reconstructed. The mean attenuation value of CM was measured at eight positions in the pulmonary trunk and pulmonary arteries. Signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR) were calculated. Qualitative assessment of the vascular enhancement was performed independently by two experienced radiologists using a three-point scale. Mean attenuation values, image noise, CNR, and SNR of images with 50 ml CM and images with 100 ml CM were compared and mean attenuation values, image noise, CNR, and SNR in 80 kV images and 120 kV images were compared. For qualitative analysis, interobserver variability was analysed using Cohen's kappa statistics. RESULTS: The mean attenuation values in Protocol I and Protocol II were not significantly different at 80 kV (634.6±168.3 versus 537.9±146.7 HU; p=0.088) and 120 kV (482.8±127.7 versus 410.4±106.0 HU; p=0.085). The mean attenuation value at 80 kV was significantly higher than the mean attenuation value at 120 kV in Protocols I and II (p<0.001). The CNR and SNR were higher at 120 kV than at 80 kV in both protocols (p=0.000-0.019); however, there were no significant differences in the CNR and SNR between both protocols (p=0.600-0.952). Qualitative (subjective) analysis showed no statistical significant difference between Protocols I and II (p=0.524-1.000). CONCLUSION: Low tube voltage (80 kV) CTA using 50 ml CM is not inferior to CTA at 120 kV using 100 ml CM.


Computed Tomography Angiography/methods , Iodine/administration & dosage , Pulmonary Artery/diagnostic imaging , Pulmonary Embolism/diagnostic imaging , Radiation Dosage , Radiation Protection/methods , Adult , Aged , Aged, 80 and over , Contrast Media/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Radiographic Image Enhancement/methods , Radiometry , Reproducibility of Results , Sensitivity and Specificity
4.
Clin Radiol ; 71(9): 940.e1-5, 2016 Sep.
Article En | MEDLINE | ID: mdl-27076253

AIM: To investigate the association between a non-invasive cardiac output (CO) measurement and the scan delay, as derived from a test bolus injection protocol. The secondary objective was to determine which factors affect the relationship between the CO and scan delay. MATERIALS AND METHODS: Fifty-five patients referred for a contrast-enhanced (thorax-)abdomen CT examination were included in this feasibility study. A test bolus examination was performed prior to the abdominal CT. During the test bolus injection, the CO of the patient was measured using a non-invasive finger-cuff measurement. Associations were analysed using linear regression analyses. Age, gender, height, weight, and blood pressure were included as potential confounders. RESULTS: Linear regression analysis showed a negative and significant association between CO and delay. The regression formula was as follows: scan delay (seconds) = 26.8-1.6 CO (l/min), with a 95% CI between -2.3 and -1.0 (p<0.001). Weight appeared to be a confounder in this relation, and gender and blood pressure were effect modifiers. There was no interaction between scan delay and age, height and weight. CONCLUSIONS: There is a negative and significant association between the non-invasive CO measurement and the CT scan delay; however, to validate these findings a larger cohort study is needed to investigate whether the non-invasively determined scan delay is as accurate as the use of a test bolus.


Aorta/metabolism , Aortography/methods , Cardiac Output/physiology , Computed Tomography Angiography/methods , Radiographic Image Interpretation, Computer-Assisted/methods , Triiodobenzoic Acids/pharmacokinetics , Computer Simulation , Contrast Media/administration & dosage , Female , Heart Function Tests/methods , Humans , Male , Middle Aged , Models, Cardiovascular , Radiography, Abdominal/methods , Reproducibility of Results , Sensitivity and Specificity , Triiodobenzoic Acids/administration & dosage
5.
Clin Radiol ; 71(1): 64-73, 2016 Jan.
Article En | MEDLINE | ID: mdl-26541440

AIM: To determine the effect of using 80 kV tube voltage and a reduced amount of contrast medium on the image quality and radiation dose of computed tomography angiography (CTA) of the abdominal aorta. MATERIALS AND METHODS: Patients who were referred for a CTA examination of the abdominal aorta were included in this technical efficacy study. Thirty patients were divided randomly into two groups. Fifteen patients underwent a dual-energy CT (DECT) protocol (Group A). Fifteen patients were scanned with the use of an automated tube potential selection algorithm tool (Group B). In both protocols, a test bolus injection of 10 ml ioversol (350 mg iodine/ml) was used, followed by 20 ml of 1:1 saline-diluted contrast medium. Quantitative analysis comprised determination of the mean attenuation and contrast-to-noise ratio. Qualitative image analysis was performed independently by five radiologists. The estimated radiation dose in terms of CT dose index and effective dose was recorded and compared with a standard 120 kV protocol. RESULTS: In Group B, six patients underwent CTA at 80 kV, seven patients underwent CTA at 100 kV and two patients underwent CTA at 120 kV. The mean contrast-enhancement values of Group A (80 kV) and the 80 kV subgroup of Group B were 16.5% and 27.6% higher compared to the 100 kV subgroup of Group B, these differences were, however, not significant. There were no significant differences in mean image quality between groups. In patients undergoing CTA at 80 kV the effective dose decreased by up to 51.3% compared to a conventional 120 kV CTA protocol. CONCLUSIONS: The findings of this study support the hypothesis that 80 kV in CTA of the abdominal aorta can reliably be used with only 30 ml contrast medium in total and a 50% reduction in radiation dose. The overall image quality was diagnostically adequate; however, it appeared to be suboptimal in patients with a BMI above 28 kg/m(2).


Angiography/methods , Aorta, Abdominal/diagnostic imaging , Contrast Media/administration & dosage , Radiation Dosage , Tomography, X-Ray Computed/methods , Triiodobenzoic Acids/administration & dosage , Aged , Algorithms , Body Mass Index , Female , Humans , Male , Prospective Studies , Radiographic Image Interpretation, Computer-Assisted , Software
6.
Clin Cancer Res ; 6(3): 942-8, 2000 Mar.
Article En | MEDLINE | ID: mdl-10741719

In this study, we show that an inhibitor of glycosphin-golipid biosynthesis, D,L-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), increases the chemosensitivity of neuroblastoma tumor cells for Taxol and vincristine. At noneffective low doses of Taxol or vincristine, the addition of a noneffective dose of PDMP resulted in 70% cytotoxicity, indicating synergy. Such an effect was not observed for etoposide (VP16). PDMP caused an early (6 h) increase in ceramide (Cer) levels, but the excess Cer was metabolically removed in the long-term (96 h). However, upon incubation with PDMP in combination with Taxol, but not with etoposide, Cer levels remained elevated at 96 h. These results suggest that neuroblastoma cells are normally able to metabolically remove excess Cer, but lose this capacity upon exposure to microtubule modulating anticancer agents (Taxol or vincristine). In addition, PDMP treatment resulted in a decreased efflux of [14C]Taxol and [3H]vincristine from neuroblastoma cells, similar to treatment with PSC833 or MK571, suggesting an effect of PDMP on the transporter proteins P-glycoprotein and/or multidrug resistance protein. PDMP did not further reduce [14C]Taxol or [3H]vincristine efflux in PSC833-treated cells, although it did further diminish cell survival under these conditions. We conclude that a combined administration of nontoxic concentrations of PDMP and either Taxol or vincristine results in highly sensitized neuroblastoma cells. This appears to involve a sustained elevation of Cer levels, possibly in concert with increased drug accumulation.


Antineoplastic Agents/pharmacology , Morpholines/pharmacology , Neuroblastoma/prevention & control , Paclitaxel/pharmacology , Vincristine/pharmacology , Animals , Cell Survival/drug effects , Ceramides/metabolism , Cyclosporins/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Neuroblastoma/pathology , Paclitaxel/metabolism , Tumor Cells, Cultured , Vincristine/metabolism
7.
Int J Cancer ; 82(1): 92-7, 1999 Jul 02.
Article En | MEDLINE | ID: mdl-10360826

Bone marrow depression is a common feature in hematological malignancies or other bone marrow-involving cancers. The mechanism of this hemopoietic suppression resulting in pancytopenia and especially anemia has not been elucidated. Gangliosides can be shed by cancer cells. Therefore, we investigated the effects of exogenously added gangliosides on erythropoiesis in a human and murine in vitro system. A dose-dependent inhibition of murine colony-forming-unit-erythroid (CFU-E) and burst-forming-unit-erythroid (BFU-E) colony growth was observed. Furthermore the maturation of BFU-Es into CFU-Es was inhibited. The inhibition by gangliosides was not abolished by increasing the dose of erythropoietin (10 U/ml). FACS-analysis studies with human CD34+ cells cultured with gangliosides (GM3), erythropoietin (EPO) and stem cell factor (SCF) demonstrated a strong inhibition on cell growth. This resulted in a significantly higher percentage of immature cells (CD34+/GpA-, 24% vs. 3%), and a lower percentage of mature erythroid cells (CD34-/GpA+, 36% vs. 89%). Under these circumstances the effects on erythroid cell growth were much higher than on other cell lineages. The inhibitory effect of gangliosides isolated from acute lymphoblastic leukemic patients on in vitro erythropoiesis suggests that in vivo hemopoietic suppression might have its origin in the gangliosides present and probably shed by the malignant cells in the microenvironment and plasma. Our results show that gangliosides inhibit erythropoiesis in vitro at several stages of development, by a mechanism involving modulation of the maturation of erythroid cells.


Bone Marrow/physiopathology , Erythropoiesis/drug effects , Gangliosides/pharmacology , Leukemia/blood , Animals , Antigens, CD34/analysis , Erythroid Precursor Cells/physiology , Erythropoietin/pharmacology , Female , Humans , Mice , Mice, Inbred C57BL
8.
Cancer Res ; 58(21): 4840-4, 1998 Nov 01.
Article En | MEDLINE | ID: mdl-9809988

Hemopoiesis is disturbed in bone marrow-involving cancers like leukemia and neuroblastoma. Shedding of gangliosides by tumor cells may contribute to this tumor-induced bone marrow suppression. We studied in vitro the inhibitory effects of murine neuroblastoma cells (Neuro-2a and C1300) and their gangliosides on hemopoiesis using normal murine hemopoietic progenitor colony-forming assays. Transwell cultured neuroblastoma cells showed a dose-dependent inhibition on hemopoiesis, indicating that a soluble factor was responsible for this effect. Furthermore, the supernatant of Neuro-2a cultured cells inhibited hemopoietic proliferation and differentiation. To determine whether the inhibitory effect was indeed due to shed gangliosides and not, for instance, caused by cytokines, the effect of DL-threo-1 -phenyl-2-decanoylamino-3-morpholino-1-propanol (DL-PDMP) on Neuro-2a cells was studied. DL-PDMP is a potent inhibitor of glucosylceramide synthase, resulting in inhibition of the synthesis and shedding of gangliosides. The initially observed inhibitory effect of supernatant of Neuro-2a cells was abrogated by culturing these cells for 3 days in the presence of 10 microM DL-PDMP. Moreover, gangliosides isolated from Neuro-2a cell membranes inhibited hemopoietic growth. To determine whether the described phenomena in vitro are a reflection of bone marrow suppression occurring in vivo, gangliosides isolated from plasma of neuroblastoma patients were tested for their effects on human hemopoietic progenitor colony-forming assays. These human neuroblastoma-derived gangliosides inhibited normal erythropoiesis (colony-forming unit-erythroid/burst-forming unit-erythroid) and myelopoiesis (colony-forming unit-granulocyte/macrophage) to a higher extent compared with gangliosides isolated from control plasma. Altogether these results suggest that gangliosides shed by neuroblastoma cells inhibit hemopoiesis and may contribute to the observed bone marrow depression in neuroblastoma patients.


Gangliosides/pharmacology , Hematopoiesis/drug effects , Neuroblastoma/physiopathology , Animals , Female , G(M3) Ganglioside/pharmacology , Humans , Mice , Mice, Inbred C57BL
9.
Anticancer Drug Des ; 13(4): 291-305, 1998 Jun.
Article En | MEDLINE | ID: mdl-9627669

Toxicities of 5-fluoro-2'-deoxyuridine (FUdR) and its liposome incorporated dipalmitoyl derivative (FUdR-dipalmitate) to mouse bone marrow, spleen, liver and ileum were compared after treatment for 6 consecutive days. The applied doses of the two formulations, which were shown earlier to have equal antitumor activity in mouse tumor models, were 600 and 2 mumol/kg respectively. When applied in these doses, toxicity to the hemopoietic system, measured as a decreases in progenitor and precursor cells of the erythroid and granuloid/macrophage lineage in bone marrow and spleen, was more severe for FUdR than for liposomal FUdR-dipalmitate. In the liver, mitotic figures, as indicators of cell division, were absent for both drugs while in control livers the number of cells in mitosis was approximately 2%. Toxicity to the ileum was more severe for liposomal FUdR-dipalmitate than for FUdR and was manifested by granulocyte infiltration, the presence of cell debris, loss of columnar epithelial cells and enlarged nuclei with prominent nucleoli in these cells. Thus, by prolonging the retention time of FUdR in vivo, using liposomes as a vehicle and FUdR-dipalmitate as a lipophilic prodrug, the dose-limiting toxicity appears to shift from bone marrow to the gastrointestinal tract.


Antineoplastic Agents/toxicity , Floxuridine/analogs & derivatives , Prodrugs/toxicity , Animals , Antineoplastic Agents/administration & dosage , Body Weight/drug effects , Bone Marrow/drug effects , Drug Carriers , Female , Floxuridine/administration & dosage , Floxuridine/toxicity , Ileum/drug effects , Ileum/pathology , Injections, Intraperitoneal , Injections, Intravenous , Liposomes , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Prodrugs/administration & dosage , Spleen/drug effects , Spleen/pathology
10.
Blood ; 91(9): 3222-9, 1998 May 01.
Article En | MEDLINE | ID: mdl-9558377

We investigated how in vivo effects of single hematopoietic cytokines change if given in combination for a prolonged time. Mice were treated with every combination of recombinant human (rh) erythropoietin (EPO), rh granulocyte colony-stimulating factor (G-CSF), recombinant rat (rr) stem cell factor (SCF), and rh interleukin (IL)-11 by continuous infusion over 7 days (full factorial design with three dose levels for each cytokine). Burst-forming unit-erythroid (BFU-E), colony-forming unit-erythroid (CFU-E), and colony-forming unit-granulocyte-macrophage (CFU-GM) were determined in bone marrow and spleen, reticulocytes, hematocrit, granulocytes, and thrombocytes in the peripheral blood. An analysis of variance (ANOVA) and multiple comparison of means was used to evaluate the data. For several cell types, cytokine effects superimposed in an additive way if combined. However, in a large number of circumstances, nonadditive pairwise interactions were found. They differed in type and magnitude involving high-dose saturation, high-dose antagonistic effects, and even effect reversals (qualitative interactions). Hence, in general, it was not possible to foresee the combination effects on the basis of existing knowledge of single effects. On the other hand, the cytokine network was robust and no system hazards were observed under multiple cytokine combinations. The results illustrate that the cytokine network has nonlinear dynamic properties in vivo with dose-response characteristics of one cytokine being continuously modified by other cytokines.


Erythropoietin/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoiesis/drug effects , Interleukin-11/administration & dosage , Stem Cell Factor/administration & dosage , Animals , Drug Interactions , Drug Synergism , Erythropoiesis/drug effects , Female , Leukopoiesis/drug effects , Mice , Mice, Inbred C57BL
11.
Radiat Res ; 147(6): 680-5, 1997 Jun.
Article En | MEDLINE | ID: mdl-9189165

The effect of stem cell factor (SCF) given at 24, 12 and 2 h before either 5-fluorouracil (5-FU) or total-body irradiation (TBI) was investigated on a range of bone marrow hemopoietic cell subsets that included primitive stem cells capable of long-term repopulation in bone marrow transplant (BMT) recipients. At 24 h after treatment, the femoral content of transient and permanent repopulating stem cell subsets was assessed from the frequency of early- and late-developing cobblestone area-forming cells (CAFCs) growing in stroma-associated cultures. At this time untreated 3 x 10(6) congenically marked donor bone marrow cells (B6-Gpi-Ia-->B6-Gpi-Ib) were transplanted and the level of erythroid engraftment was followed over 1 year. Analysis of the frequencies of CAFCs in host bone marrow after treatment with SCF demonstrated a remarkable increase in the number of early-developing CAFC subsets by about 10-fold. At the same time SCF conferred a sensitization of these subsets after treatment with 5-FU, which indicated an enhanced proliferative activity. The SCF-induced increase in the number of progenitor cells, however, was the more dominant process in the irradiated animals, resulting in less overall depletion of CAFCs. These contrasting results provide an explanation for the sensitization by SCF of 5-FU-induced lethality and its converse protection against radiation-induced lethality as reported by others. Nevertheless, the number of the more primitive CAFC subsets appearing at 28 and 35 days in culture and their sensitivity to 5-FU or radiation remained unaffected by this short SCF treatment. The number of CAFCs that remained in the bone marrow largely predicted the subsequent patterns of donor marrow engraftment in the treated BMT recipients: SCF enhanced short-term engraftment after treatment with 5-FU while it reduced the need for short-term engraftment after irradiation. Only irradiation afforded long-term engraftment through depletion of primitive host stem cells, and this was moderately improved by prior treatment with SCF.


Bone Marrow Transplantation , Fluorouracil/pharmacology , Hematopoietic Stem Cells/drug effects , Stem Cell Factor/pharmacology , Whole-Body Irradiation , Animals , Male , Mice , Mice, Inbred C57BL
12.
Clin Chem ; 43(4): 644-8, 1997 Apr.
Article En | MEDLINE | ID: mdl-9105267

Hb A1c is the analyte of choice for monitoring metabolic control in patients with diabetes mellitus. Here we present a new analytical technique for measuring Hb A1c, capillary electrophoresis. The Hb A1c determination is not influenced by the labile Hb A1c fraction or by carbamylated or acetylated hemoglobin derivatives. Also, hemoglobin variants (Hb F, Hb S, and Hb C) do not interfere. This new application of capillary electrophoresis seems to be a valuable analytical tool for measuring Hb A1c in the clinical laboratory.


Blood Protein Electrophoresis/methods , Diabetes Mellitus/blood , Electrophoresis, Capillary/methods , Glycated Hemoglobin/analysis , Electrophoresis, Capillary/statistics & numerical data , Humans , Reproducibility of Results , Sensitivity and Specificity
13.
Blood ; 89(5): 1543-50, 1997 Mar 01.
Article En | MEDLINE | ID: mdl-9057635

We have quantified the frequency and proliferation of five subsets of primitive hematopoietic cells, using the cobblestone area forming cell (CAFC) assay, in marrow of five strains of mice with lifespans ranging from about 500 to 800 days. Stem cell characteristics were determined in young (6 weeks) and old (12 months) mice. We report striking effects of both intrinsic strain lifespan and individual mouse age on stem cell populations. First, the relative and absolute numbers of the most primitive stem cell subsets was threefold to fourfold higher in old than in young mice. Second, a considerable strain-to-strain variation in the number of primitive cells was observed: when absolute frequencies were calculated, there was a trend for longer lifespan to be correlated with a larger stem cell pool. Third, stem cells from old mice had a far lower cycling activity than cells from young mice. However, this was highly strain dependent: short-lived C3H/He and CBA/J mice showed a stronger reduction in cycling activity during aging than long-lived C57BL/6 mice. Finally, a significant negative correlation was demonstrated in young mice between maximal lifespan and proliferative activity. These data show that aging has a major impact on the frequency and cell-cycle kinetics of primitive hematopoietic cell compartments. In addition, the observation that cycling activity of stem cells is related to the maximal lifespan of the mouse strain may open ways to identify the genetic mechanisms of both strain- and age-dependent variation in the structure of primitive hematopoietic cell compartments.


Aging/blood , Hematopoietic Stem Cells/pathology , Mice, Inbred Strains , Aging/pathology , Animals , Cell Count , Cell Division , Mice , Species Specificity
14.
Leuk Lymphoma ; 22(5-6): 385-94, 1996 Aug.
Article En | MEDLINE | ID: mdl-8882951

Hemopoietic cell amplification in vivo is regulated by various mechanisms which appear to be under control of many hemopoietic growth factors. Quiescent stem cells can be activated into cell cycle, dividing progenitor cells can reduce their cycle time, the differentiation velocity (i.e. transit-time) can be manipulated, apoptosis can be prevented, and finally, at least in the murine system, migration of cells between the microenvironments in marrow and spleen may take place. Perturbations of any of the parameters by which these mechanisms are defined, will affect in vivo blood cell production. In this review the consequences of these perturbations, and the role of growth factors herein, are discussed. These fundamental aspects of the regulation of hemopoiesis are illustrated with recent data showing the synergistic, redundant and pleiotropic effects of SCF, IL-11, EPO and G-CSF on the in vivo formation of erythrocytes. Given the overwhelming number of growth factor-related studies that are now appearing, a re-evaluation of data, available in the literature, in the context of the mechanistic approach of growth factor-dependent hemopoiesis which is presented in this paper, seems to be useful and warranted.


Cytokines/physiology , Erythropoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Bone Marrow Cells , Cell Cycle , Cell Differentiation , Cell Division , Cytokines/pharmacology , Erythrocytes/cytology , Erythrocytes/physiology , Erythropoiesis/drug effects , Growth Substances/pharmacology , Growth Substances/physiology , Hematopoietic Stem Cells/drug effects , Humans , Kinetics , Models, Biological , Spleen/cytology , Time Factors
15.
Blood ; 87(11): 4581-8, 1996 Jun 01.
Article En | MEDLINE | ID: mdl-8639826

The aim of this study was to expand the primitive and committed hematopoietic cell compartments in vivo in order to confer resistance of the blood cell forming system against the cytotoxic, cell cycle specific drug 5-fluorouracil (5-FU). Possible chemoprotective effects of such a pretreatment could result from increased numbers of hematopoietic cells, present before 5-FU administration. In addition, we hypothesized that an enhanced number of primitive and progenitor calls would result in a reduced cycling activity, ie, 5-FU sensitivity, of these same cells, due to normal physiological feedback loops. Administration of stem cell factor (SCF) plus interleukin-11 (IL-11) to mice was shown to result in expansion of the various immature cell compartments in marrow and, in particular, spleen. The total body content of the primitive cobblestone area forming cells (CAFC)-day 28 was increased to 140%, whereas the more committed cells (CAFC-day 7, erythroid and granuloid progenitors) were increased to 500%. This in vivo expansion resulted in a decreased 5-FU sensitivity of the hematopoietic system. In particular, mice that had received 5-FU 24 hours after discontinuation of growth factor pretreatment showed significantly less toxicity of committed cell stages. Compared with mice not pretreated, it appeared that in pretreated mice, 24 hours after 5-FU administration, the absolute number, but also the fraction of surviving CAFC, was much higher in both marrow and spleen. This was caused by a decrease in the cycling activity of all primitive cell subsets. To explore the possible use of this finding in a chemotherapeutic setting, we determined the interval between two subsequent doses of 5-FU (160 mg/kg) that was required to prevent drug-induced mortality. When control mice received a second dose of 5-FU 7, 10, or 14 days after the first, respectively 0%, 20%, and 80% survived. In contrast, 40% and 100% of mice that received SCF + IL-11 before the first dose of 5-FU, survived a second dose of 5-FU given respectively after 7 or 10 days. To assess whether chemoprotection in this setting could be ascribed to protection of the hematopoietic system, we transplanted a high number of normal bone marrow cells (sufficient to compensate for any hematopoietic deficiency) to normal and pretreated mice after they had been administered 2 doses of 5-FU, given 7 days apart. Bone marrow transplantation (BMT) could only rescue 50% of mice not pretreated, showing that a significant part of the mortality was because of nonhematologic toxicity. However, a BMT given to growth factor pretreated mice saved all mice, indicating that in this setting SCF + IL-11 had additional protective effects on cell systems other than hematopoiesis. In conclusion, our study showed fundamental knowledge about the behavior of primitive cells in vivo and has shown that manipulation of these and other cell compartments with appropriate growth factors may confer resistance against cytotoxic drugs.


Fluorouracil/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Interleukin-11/pharmacology , Stem Cell Factor/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow Cells , Cell Differentiation/drug effects , Drug Evaluation, Preclinical , Drug Resistance , Feedback , Female , Fluorouracil/toxicity , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Rats , Recombinant Proteins/pharmacology
16.
Blood ; 86(8): 2986-92, 1995 Oct 15.
Article En | MEDLINE | ID: mdl-7579391

Because of the complexity of appropriate stem cell assays, little information on the in vivo regulation of murine stem cell biology or stemmatopoiesis is available. It is unknown whether and how in vivo the primitive hematopoietic stem cell compartment is affected during a continued increased production of mature blood cells. In this study, we present data showing that prolonged (3 weeks) administration of granulocyte colony-stimulating factor (G-CSF), which is a major regulator of mature granulocyte production, has a substantial impact on both the size and the location of various stem cell subset pools in mice. We have used the novel cobblestone area forming cell (CAFC) assay to assess the effects of G-CSF on the stem cell compartment (CAFC days 7, 14, 21, and 28). In marrow, in which normally 99% of the total number of stem cells can be found, G-CSF induced a severe depletion of particularly the most primitive stem cells to 5% to 10% of normal values. The response after 7 days of G-CSF treatment was an increased amplification between CAFC day 14 and 7. However, this response occurred at the expense of the number of CAFC day 14. It is likely that the resulting gap of CAFC day 14 cell numbers was subsequently replenished from the more primitive CAFC day 21 and 28 compartments, because these cell numbers remained low during the entire treatment period. In the spleen, the number of stem cells increased, likely caused by a migration from the marrow via the blood, leading to an accumulation in the spleen. The increased number of stem cells in the spleen overcompensated for the loss in the marrow. When total body (marrow and spleen) stem cell numbers were calculated, it appeared that a continued increased production of mature granulocytes resulted in the establishment of a higher, new steady state of the stem cell compartment; most committed stem cells (CAFC day 7) were increased threefold, CAFC day 14 were increased 2.3-fold, CAFC-day 21 were increased 1.8-fold, and the most primitive stem cells evaluated, CAFC day 28, were not different from normal, although now 95% of these cells were located in the spleen. Four weeks after discontinuation of the G-CSF treatment, the stem cell reserve in the spleen had returned to a normal level, whereas stem cell numbers in marrow had recovered to values above normal. This study shows that the primitive stem cell compartment is seriously perturbed during an increased stimulation of the production of mature blood cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Erythropoiesis/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Animals , Bone Marrow/drug effects , Bone Marrow Cells , Cell Count , Cell Cycle/drug effects , Cell Division/drug effects , Feedback , Female , Hematopoiesis, Extramedullary/drug effects , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/drug effects
17.
Br J Haematol ; 90(4): 783-90, 1995 Aug.
Article En | MEDLINE | ID: mdl-7545423

In this study we evaluated the in vivo effects of interleukin-11 (IL-11) and stem cell factor (SCF), in combination with erythropoietin (EPO) on murine erythropoiesis. Mice were treated for 7 d with IL-11. SCF and EPO, each at three dose levels. In total, 27 different dose combinations were tested. IL-11 as well as SCF could only marginally stimulate erythroid progenitor cell numbers, but IL-11 in combination with SCF was able to increase BFU-E and CFU-E numbers 4-fold, in the absence of exogenous EPO. This resulted in an increased reticulocyte count. In contrast with the stimulatory effect on immature erythroid cell stages, IL-11 treatment induced a mild anaemia, which probably resulted from a plasma volume expansion. The additional treatment with EPO resulted in strong synergistic effects on CFU-E numbers. The combination of high-dose IL-11 and high-dose SCF was able to increase the overall efficiency of EPO-induced erythroid amplification, which was reflected by a left-shift of the in vivo EPO dose-response curve. The stimulating effects of IL-11 and SCF were further demonstrated when the effects on the reticulocyte count of a single high-dose EPO injection were assessed in normal and SCF+IL-11 treated mice. Whereas a single EPO dose increased the reticulocyte count by a factor of 3, IL-11 + SCF pretreatment increased this to a factor of 7. This study shows that in vivo SCF and IL-11 are important modulators of red blood cell production. First, these factors probably increase the input from the stem cell compartment into the erythroid lineage, where subsequently EPO is required for further amplification. Additionally, however, IL-11 and SCF increase the overall efficiency of EPO-induced amplification, probably due to a stimulatory effect on late-stage erythroid cells and to a redistribution of cells from marrow to spleen.


Erythroid Precursor Cells/physiology , Erythropoiesis/physiology , Erythropoietin/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Interleukin-11/pharmacology , Animals , Erythropoietin/administration & dosage , Female , Hematopoietic Cell Growth Factors/administration & dosage , Interleukin-11/administration & dosage , Mice , Mice, Inbred C57BL , Reticulocytes/physiology , Stem Cell Factor
18.
Exp Hematol ; 23(4): 369-75, 1995 Apr.
Article En | MEDLINE | ID: mdl-7895784

The in vivo mechanism of hematopoietic growth factor-induced cell multiplication is in debate. Several options can be examined: 1) growth factors can reduce the cycling time of their dividing target cells, 2) growth factors can add extra cell divisions within the differentiation pathway, 3) the combination of the first two possibilities, and 4) growth factors can prevent premature cell death (apoptosis) from occurring in the absence of the stimulating factor. We studied these options in vitro and in vivo in the murine erythroid pathway. Results from in vitro cultures of purified splenic colony-forming units-erythroid (CFU-E), with and without erythropoietin (Epo), and in vivo Epo treatments of thiamphenicol (TAP)-pretreated mice showed neither reduction in cycle times nor addition of extra cell divisions in the differentiating erythroid lineage. The phenomenon of apoptosis was demonstrated as time- and Epo-dependent in vitro with electrophoretic (DNA-ladder), flow-cytometric (subdiploid cells), and morphologic (fragmented nuclei) methods applied on CFU-E. A high dose of Epo administered to mice caused a rapid transient rise in the number of CFU-E to 350% of normal. Early erythroblasts also increased, whereas burst-forming unit-erythroid (BFU-E) numbers did not change. Our results favor a mechanism in which Epo acts as a survival factor for early erythroid cells (CFU-E and early erythroblasts) in vitro, as well as in vivo, preventing apoptosis.


Erythropoiesis/drug effects , Erythropoietin/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL
19.
Stem Cells ; 13(1): 65-76, 1995 Jan.
Article En | MEDLINE | ID: mdl-7536513

The aim of this study was to determine how stem cell factor (SCF) modifies hemopoietic cell production. First we determined the effects of a prolonged SCF administration on murine hemopoiesis and analyzed the results by a mathematical simulation model of hemopoiesis in order to explain the data. Subsequently we investigated the effects of simultaneous coadministration of SCF+erythropoietin (Epo), to see how effects of early and late cytokines superimpose. SCF administration during 14 days induced a proliferative wave through the hemopoietic system; colony forming units-granulocyte macrophage (CFU-GM), burst forming units-erythroid (BFU-E) and colony forming units erythroid (CFU-E) were the first to be augmented, followed by their respective progeny, ultimately leading to increased blood cell numbers. Despite continued treatment most cell numbers returned to normal values in 14 days, colony forming units-spleen (CFU-S), however, remained elevated. This wave pattern could be explained within the framework of a previously established mathematical model of hemopoiesis, if it was assumed that SCF affected the cycling status of primitive cells and if regulatory feedback loops of erythroid and myeloid progenitors on these cells were also allowed. Simultaneous SCF and Epo administration led to synergistic effects on CFU-E numbers and hematocrit values at moderate Epo doses. At high Epo doses, however, this was less pronounced. We conclude that SCF increases the input into committed hemopoietic lineages, where late acting cytokines can induce further amplification.


Erythropoietin/administration & dosage , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/administration & dosage , Animals , Bone Marrow Cells , Colony-Forming Units Assay , Drug Administration Schedule , Female , Granulocytes/cytology , Mice , Mice, Inbred C57BL , Spleen/cytology , Stem Cell Factor
20.
Cancer Res ; 55(2): 324-9, 1995 Jan 15.
Article En | MEDLINE | ID: mdl-7529132

In this study, we determined in vivo interactions between hemopoietic growth factors and etoposide (VP-16) to assess whether normal blood cell production could be maintained during chemotherapy if hemopoietic growth factors were simultaneously administered. Groups of mice were treated for 7 consecutive days with four different doses of VP-16 in combination with three different doses of erythropoietin (EPO) or granulocyte colony-stimulating factor (G-CSF). In total, 12 combinations of VP-16 plus EPO and 12 combinations of VP-16 plus G-CSF were thus evaluated. Intricate dose-response surfaces of the effects of the different treatments on colony-forming units-erythroid, reticulocytes, hematocrit, colony-forming units-granulocyte/macrophage, and absolute neutrophil count were obtained, which revealed that: (a) simultaneous EPO administration was able to maintain reticulocyte production and to protect mice from VP-16 induced anemia; (b) simultaneous G-CSF administration was able to maintain granulocyte production and to protect mice from VP-16 induced neutropenia; (c) VP-16 dose escalation was feasible when EPO or G-CSF were simultaneously administered; and (d) no increased myelotoxicity on erythroid or granuloid progenitors was observed when EPO or G-CSF was simultaneously administered with VP-16. These results suggest that in vivo either individual hemopoietic progenitors can become resistant against VP-16-induced cell death by appropriate simultaneous growth factor administration or that the loss of overall cell amplification, induced by VP-16, can be compensated by extra amplification of surviving progenitors. Furthermore, these data indicate that a strict separation in time of cytostatic drug and growth factor treatment is not necessarily the optimal schedule with respect to the reduction of hemotoxicity.


Anemia/prevention & control , Erythropoietin/administration & dosage , Etoposide/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cells/drug effects , Neutropenia/prevention & control , Anemia/chemically induced , Animals , Bone Marrow/drug effects , Bone Marrow Cells , Dose-Response Relationship, Drug , Erythroid Precursor Cells/drug effects , Etoposide/adverse effects , Female , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neutropenia/chemically induced , Regression Analysis , Spleen/cytology , Spleen/drug effects
...