Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(20): eadk9076, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38748792

ABSTRACT

Acute myeloid leukemia (AML) driven by the activation of EVI1 due to chromosome 3q26/MECOM rearrangements is incurable. Because transcription factors such as EVI1 are notoriously hard to target, insight into the mechanism by which EVI1 drives myeloid transformation could provide alternative avenues for therapy. Applying protein folding predictions combined with proteomics technologies, we demonstrate that interaction of EVI1 with CTBP1 and CTBP2 via a single PLDLS motif is indispensable for leukemic transformation. A 4× PLDLS repeat construct outcompetes binding of EVI1 to CTBP1 and CTBP2 and inhibits proliferation of 3q26/MECOM rearranged AML in vitro and in xenotransplant models. This proof-of-concept study opens the possibility to target one of the most incurable forms of AML with specific EVI1-CTBP inhibitors. This has important implications for other tumor types with aberrant expression of EVI1 and for cancers transformed by different CTBP-dependent oncogenic transcription factors.


Subject(s)
Alcohol Oxidoreductases , DNA-Binding Proteins , Leukemia, Myeloid, Acute , MDS1 and EVI1 Complex Locus Protein , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , MDS1 and EVI1 Complex Locus Protein/metabolism , MDS1 and EVI1 Complex Locus Protein/genetics , Alcohol Oxidoreductases/metabolism , Alcohol Oxidoreductases/genetics , Humans , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/genetics , Protein Binding , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
2.
Blood ; 128(25): 2949-2959, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27733356

ABSTRACT

To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Stem Cell Niche , Tissue Scaffolds/chemistry , Xenograft Model Antitumor Assays , Animals , Cell Self Renewal , Cell Separation , Clone Cells , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/cytology , Mice , Phenotype , Stromal Cells/pathology
4.
Methods Mol Biol ; 1416: 225-32, 2016.
Article in English | MEDLINE | ID: mdl-27236674

ABSTRACT

Large animal models are an important preclinical tool for the evaluation of new interventions and their translation into clinical practice. The pig is a widely used animal model in multiple clinical fields, such as cardiology and orthopedics, and has been at the forefront of testing new therapeutics, including cell-based therapies. In the clinic, mesenchymal stem cells (MSCs) are used autologously, therefore isolated, and administrated into the same patient. For successful clinical translation of autologous approaches, the porcine model needs to test MSC in a similar manner. Since a limited number of MSCs can be isolated directly from the bone marrow, culturing techniques are needed to expand the population in vitro prior to therapeutic application. Here, we describe a protocol specifically tailored for the isolation and propagation of porcine-derived bone marrow MSCs.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mesenchymal Stem Cell Transplantation , Sus scrofa , Swine , Transplantation, Autologous
5.
Haematologica ; 101(5): 616-25, 2016 05.
Article in English | MEDLINE | ID: mdl-26858358

ABSTRACT

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we transduced T cells from healthy donors and multiple myeloma patients. We then evaluated the preclinical efficacy and safety of the transduced T cells. Irrespective of the donor and antibody sequence, CD38-chimeric antigen receptor-transduced T cells proliferated, produced inflammatory cytokines and effectively lysed malignant cell lines and primary malignant cells from patients with acute myeloid leukemia and multi-drug resistant multiple myeloma in a cell-dose, and CD38-dependent manner, despite becoming CD38-negative during culture. CD38-chimeric antigen receptor-transduced T cells also displayed significant anti-tumor effects in a xenotransplant model, in which multiple myeloma tumors were grown in a human bone marrow-like microenvironment. CD38-chimeric antigen receptor-transduced T cells also appeared to lyse the CD38(+) fractions of CD34(+) hematopoietic progenitor cells, monocytes, natural killer cells, and to a lesser extent T and B cells but did not inhibit the outgrowth of progenitor cells into various myeloid lineages and, furthermore, were effectively controllable with a caspase-9-based suicide gene. These results signify the potential importance of CD38-chimeric antigen receptor-transduced T cells as therapeutic tools for CD38(+) malignancies and warrant further efforts to diminish the undesired effects of this immunotherapy using appropriate strategies.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Immunotherapy , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Animals , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Flow Cytometry , Gene Expression , Gene Transfer Techniques , Genes, Transgenic, Suicide , Hematopoietic Stem Cells/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Burden/genetics , Tumor Burden/immunology
6.
Cancer Immunol Immunother ; 64(8): 951-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25920521

ABSTRACT

Immunotherapy with allogeneic natural killer (NK) cells offers therapeutic perspectives for multiple myeloma patients. Here, we aimed to refine NK cell therapy by evaluation of the relevance of HLA-class I and HLA-E for NK anti-myeloma reactivity. We show that HLA-class I was strongly expressed on the surface of patient-derived myeloma cells and on myeloma cell lines. HLA-E was highly expressed by primary myeloma cells but only marginally by cell lines. HLA-E(low) expression on U266 cells observed in vitro was strongly upregulated after in vivo (bone marrow) growth in RAG-2(-/-) γc(-/-) mice, suggesting that in vitro HLA-E levels poorly predict the in vivo situation. Concurrent analysis of inhibitory receptors (KIR2DL1, KIR2DL2/3, KIR3DL1 and NKG2A) and NK cell degranulation upon co-culture with myeloma cells revealed that KIR-ligand-mismatched NK cells degranulate more than matched subsets and that HLA-E abrogates degranulation of NKG2A+ subsets. Inhibition by HLA-class I and HLA-E was also observed with IL-2-activated NK cells and at low oxygen levels (0.6 %) mimicking hypoxic bone marrow niches where myeloma cells preferentially reside. Our study demonstrates that NKG2A-negative, KIR-ligand-mismatched NK cells are the most potent subset for clinical application. We envision that infusion of high numbers of this subclass will enhance clinical efficacy.


Subject(s)
Cell Separation/methods , Histocompatibility Antigens Class I/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , NK Cell Lectin-Like Receptor Subfamily C/immunology , Animals , Cell Degranulation , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , Interleukin-2/immunology , Mice , Mice, Knockout , Multiple Myeloma/immunology , Neoplasm Transplantation , Oxygen/metabolism , HLA-E Antigens
7.
Clin Cancer Res ; 21(12): 2802-10, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25398450

ABSTRACT

PURPOSE: Novel therapeutic agents have significantly improved the survival of patients with multiple myeloma. Nonetheless, the prognosis of patients with multiple myeloma who become refractory to the novel agents lenalidomide and bortezomib is very poor, indicating the urgent need for new therapeutic options for these patients. The human CD38 monoclonal antibody daratumumab is being evaluated as a novel therapy for multiple myeloma. Prompted with the encouraging results of ongoing clinical phase I/II trials, we now addressed the potential value of daratumumab alone or in combination with lenalidomide or bortezomib for the treatment of lenalidomide- and bortezomib-refractory patients. EXPERIMENTAL DESIGN: In ex vivo assays, mainly evaluating antibody-dependent cell-mediated cytotoxicity, and in an in vivo xenograft mouse model, we evaluated daratumumab alone or in combination with lenalidomide or bortezomib as a potential therapy for lenalidomide- and bortezomib-refractory multiple myeloma patients. RESULTS: Daratumumab induced significant lysis of lenalidomide/bortezomib-resistant multiple myeloma cell lines and of primary multiple myeloma cells in the bone marrow mononuclear cells derived from lenalidomide- and/or bortezomib-refractory patients. In these assays, lenalidomide but not bortezomib, synergistically enhanced daratumumab-mediated multiple myeloma lysis through activation of natural killer cells. Finally, in an in vivo xenograft model, only the combination of daratumumab with lenalidomide effectively reduced the tumorigenic growth of primary multiple myeloma cells from a lenalidomide- and bortezomib-refractory patient. CONCLUSIONS: Our results provide the first preclinical evidence for the benefit of daratumumab plus lenalidomide combination for lenalidomide- and bortezomib-refractory patients.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Bortezomib/pharmacology , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Thalidomide/analogs & derivatives , Adult , Aged , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Bortezomib/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , Immunotherapy , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , Molecular Targeted Therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Thalidomide/administration & dosage , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
8.
Cardiovasc Res ; 99(1): 83-91, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23554461

ABSTRACT

AIMS: One of the main limitations for an effective cell therapy for the heart is the poor cell engraftment after implantation, which is partly due to a large percentage of cell death in the hostile myocardium. In the present study, we investigated the utilization of necrostatin-1 (Nec-1) as a possible attenuator of cell death in cardiomyocyte progenitor cells (CMPCs). METHODS AND RESULTS: In a mouse model of myocardial infarction, survival of CMPCs 3 days after intra-myocardial injection was 39 ± 9% higher in cells pretreated with the Nec-1 compound. However, the increase in cell number was not sustained over 28 days, and did not translate into improved cardiac function (ejection fraction %, 20.6 ± 2.1 vs. 21.4 ± 2.5 for vehicle and Nec-1-treated CMPC, respectively). Nonetheless, Nec-1 rescued CMPCs remained functionally competent. CONCLUSION: A pharmacological pretreatment approach to solely enhance cell survival on the short term does not seem to be effective strategy to improve cardiac cell therapy with CMPCs.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Myocardial Infarction/surgery , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Stem Cell Transplantation , Stem Cells/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function , Stem Cells/metabolism , Stem Cells/pathology , Stroke Volume , Time Factors , Transduction, Genetic , Transfection , Ventricular Function, Left
9.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653974

ABSTRACT

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Subject(s)
Hematopoietic Stem Cells/cytology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Stem Cell Niche/immunology , Transplantation Chimera/immunology , Tumor Microenvironment/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear Ossicles/cytology , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Mutant Strains , Neoplasm Transplantation , Osteolysis/immunology , Tissue Scaffolds , Transplantation, Heterologous
10.
Br J Haematol ; 148(3): 428-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19863541

ABSTRACT

Mesenchymal stromal cells (MSC) are potential cells for cellular therapies, in which the recruitment and migration of MSC towards injured tissue is crucial. Our data show that culture-expanded MSC from fetal lung and bone marrow, adult bone marrow and adipose tissue contained a small percentage of migrating cells in vitro, but the optimal stimulus was different. Overall, fetal lung-MSC had the highest migratory capacity. As fetal bone marrow-MSC had lower migratory potential than fetal lung-MSC, the tissue of origin may determine the migratory capacity of MSC. No additive effect in migration towards combined stimuli was observed, which suggests only one migratory MSC fraction. Interestingly, actin rearrangement and increased paxillin phosphorylation were observed in most MSC upon stromal cell-derived factor-1alpha or platelet-derived growth factor-BB stimulation, indicating that this mechanism involved in responding to migratory cues is not restricted to migratory MSC. Migratory MSC maintained differentiation and migration potential, and contained significantly less cells in S- and G2/M-phase than their non-migrating counterpart. In conclusion, our results suggest that MSC from various sources have different migratory capacities, depending on the tissue of origin. Similar to haematopoietic stem cells, cell cycle contributes to MSC migration, which offers perspectives for modulation of MSC to enhance efficacy of future cellular therapies.


Subject(s)
Fetus/cytology , Mesenchymal Stem Cells/physiology , Actins/metabolism , Adipose Tissue/cytology , Adult , Bone Marrow/embryology , Bone Marrow Cells/physiology , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Chemotaxis/physiology , Humans , Integrins/metabolism , Lung/cytology , Lung/embryology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Paxillin/metabolism , Phosphorylation , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/physiology
11.
Pediatr Cardiol ; 30(5): 699-709, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19184178

ABSTRACT

Heart failure emerges with a net loss of viable cardiomyocytes, and there is no current therapy to reverse this process to improve long-term cardiac function. Due to a change in viewpoint, that the human heart cannot be considered a terminally differentiated postmitotic organ, incapable of myocardial regeneration, a belief in a new approach for therapy evolved: regenerating the heart. Finding stem cells in the heart capable of replenishing lost cardiomyocytes became a holy grail for research. Heart stem cells were isolated and characterized, originally derived from in- or outside of the heart. Since the endogenous repair potential of the heart following injury is not sufficient, cellular therapy has been performed after myocardial infarction in clinical settings. Clinical therapies performed with autologous skeletal myoblasts, cardiomyocytes, and bone marrow, as well as the animal studies, showed improvements in cardiac function, although the clinical effects are still limited. These findings have stimulated optimism that progression of heart failure might be prevented or even reversed with cell-based therapy. For future research, it will be a challenge to isolate the most potent therapeutic cell with an intrinsic capacity to stimulate regeneration in the heart, by direct participation or by producing paracrine factors.


Subject(s)
Heart/physiology , Regeneration/physiology , Stem Cells/physiology , Bone Marrow , Heart Diseases/therapy , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cell Transplantation
12.
EuroIntervention ; 4(1): 133-8, 2008 May.
Article in English | MEDLINE | ID: mdl-19112790

ABSTRACT

AIMS: Clinical trials showed contradictory results in functional recovery after intracoronary infusion of autologous mononuclear (bone marrow) cells in patients with acute myocardial infarction. A recent study suggests that this might be related to the isolation protocol used. In The Netherlands, a comparable randomised multicentre trial (HEBE) was designed. To validate the isolation method of bone marrow and peripheral blood-derived mononuclear cells, we compared our processing protocol with methods comparable to the ASTAMI (no beneficial effect) and the REPAIR-AMI study (beneficial effect). METHODS AND RESULTS: The effect of several factors (density gradient, washing buffer and centrifugation speed) has been studied on recovery and function (migration and clonogenic capacity) of mononuclear cells. Significantly lower cell recoveries were found at a centrifugation speed of 250 g, compared to 600 or 800 g, respectively. Furthermore, washing buffer without supplemented human serum albumin and heparin resulted in significantly lower cell recovery and functional impairment as measured by clonogenic capacity. CONCLUSIONS: The results of our study justify the cell-processing protocol as applied in the HEBE trial (600 g, human serum albumin supplemented washing buffer). This protocol results in viable and functional cells of which the quantity and quality is at least comparable to a successful study like the REPAIR-AMI.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Cell Separation/standards , Leukocytes, Mononuclear/cytology , Myocardial Infarction/surgery , Sternum/cytology , Anticoagulants/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Buffers , Cardiac Surgical Procedures , Cell Movement , Centrifugation/methods , Clinical Protocols , Clinical Trials as Topic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Heparin/pharmacology , Humans , Leukocytes, Mononuclear/drug effects , Serum Albumin/pharmacology , Stem Cell Transplantation
13.
J Immunol ; 180(7): 5141-8, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354240

ABSTRACT

The number of colony forming unit-endothelial cells (CFU-EC) in human peripheral blood was found to be a biological marker for several vascular diseases. In this study, the heterogeneous composition of immune cells in the CFU-ECs was investigated. We confirmed that monocytes are essential for the formation of CFU-ECs. Also, however, CD4(+) T cells were found to be indispensable for the induction of CFU-EC colonies, mainly through cell-cell contact. By blocking or activating CD3 receptors on CD4(+) T cells or blocking MHC class II molecules on monocytes, it was shown that TCR-MHCII interactions are required for induction of CFU-EC colonies. Because the supernatant from preactivated T cells could also induce colony formation from purified monocytes, the T cell support turned out to be cytokine mediated. Gene expression analysis of the endothelial-like colonies formed by CD14(+) cells showed that colony formation is a proangiogenic differentiation and might reflect the ability of monocytes to facilitate vascularization. This in vitro study is the first to reveal the role of TCR-MHC class II interactions between T cells and monocytes and the subsequent inflammatory response as stimulus of monocytic properties that are associated with vascularization.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Lipopolysaccharide Receptors/immunology , Stem Cells/immunology , CD3 Complex/immunology , Cell Communication , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Histocompatibility Antigens Class II/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Monocytes/cytology , Monocytes/immunology , Neovascularization, Physiologic/genetics , Phenotype , Receptors, Antigen, T-Cell/immunology
14.
Exp Hematol ; 33(10): 1249-56, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16219548

ABSTRACT

OBJECTIVE: Umbilical cord blood (UCB) is considered as an attractive alternative source of hematopoietic stem cells for allogeneic stem cell transplantations in patients who lack human leukocyte antigen (HLA)-matched donors. However, the low cell dose adversely affects hematopoietic recovery and therefore limits application of UCB transplantation in adults. Transplantation of multiple UCB units could be a strategy to overcome cell dose limitations. MATERIALS AND METHODS: To investigate the effect of double cord transplantation, nonobese diabetic/severe combined immunodeficient mice were transplanted with human hematopoietic progenitor cells (CD34(+)) derived from two UCB units with HLA disparity. Human cell engraftment and donor origin was determined by flow cytometry. RESULTS: Double CB transplantation resulted in increased engraftment levels in the bone marrow and peripheral blood in comparison with recipients of a single unit. Because this effect could be due to the higher cell dose (2.10(5) vs 1.10(5) cells), double CB transplantation was compared with single units containing equal cell numbers (2.10(5)). In some cases, engraftment levels in recipients of single units containing 2.10(5) cells were significantly higher than after transplantation of 1.10(5) cells. These engraftment levels were similar to those observed after double CB transplantation. Chimerism analysis indicated that increased engraftment in recipients of two units was predominantly derived from one unit, whereas in other cases the contribution of the two units was similar. CONCLUSION: These results indicate that engraftment may be enhanced by addition of a second unrelated CB that might be attributed to a cell dose effect or due to a graft-facilitating effect.


Subject(s)
Blood Donors , Cord Blood Stem Cell Transplantation , Graft Survival/physiology , Hematopoiesis/physiology , Recovery of Function/physiology , Adult , Animals , Antigens, CD34/metabolism , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Fetal Blood/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Histocompatibility Testing , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous , Transplantation, Homologous
15.
Exp Hematol ; 31(10): 881-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14550803

ABSTRACT

OBJECTIVE: Previously, we have found that human culture-expanded fetal lung-derived mesenchymal stem cells (MSC) promote the engraftment of umbilical cord blood (UCB)-derived CD34((+)) cells. The high frequency of MSC in fetal lung allowed us to study whether this represented a biological feature of these cells or a property that was acquired during expansion in culture. MATERIALS AND METHODS: Irradiated NOD/SCID mice (n=80) were transplanted with 0.1x10(6) UCB CD34(+) cells in the presence or absence of 10(6) primary nonexpanded or culture-expanded fetal lung, liver, or BM CD45(-) cells, or with nonexpanded fetal lung liver or BM CD45(-) cells only. RESULTS: In comparison with transplantation of UCB CD34(+) cells only, cotransplantation of UCB CD34(+) cells and primary fetal lung or BM CD45(-) cells resulted in a significantly higher level of engraftment (% hCD45(+) cells) in BM, PB, and spleen. In addition, primary mesenchymal cells derived from adult BM had a similar promoting effect. The engraftment-enhancing effect was similar to that of culture-expanded fetal lung and BM MSC. Primary mesenchymal cells, but not culture-expanded MSC, were detected in recipient mice, suggesting that the primary cells were able to home and that this capacity was lost after expansion. CONCLUSIONS: These results show that primary mesenchymal cells from fetal lung and BM promote the engraftment of UCB-derived CD34(+) cells to a similar degree as culture-expanded MSC, indicating that it reflects a biological property of primary MSC that is preserved during expansion in culture.


Subject(s)
Antigens, CD34/analysis , Bone Marrow Transplantation , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Lung/cytology , Mesoderm/cytology , Animals , Cell Movement , Humans , Immunophenotyping , Liver/cytology , Mice , Mice, Nude , Mice, SCID
16.
Haematologica ; 88(8): 845-52, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12935972

ABSTRACT

BACKGROUND AND OBJECTIVES: We previously found that human fetal lung is a rich source of mesenchymal stem cells (MSC). Here we characterize and analyze the frequency and function of MSC in other second-trimester fetal tissues. DESIGN AND METHODS: Single cell suspensions of fetal bone marrow (BM), liver, lung, and spleen were made and analyzed by flow cytometry for the expression of CD90, CD105, CD166, SH3, SH4, HLA-ABC, HLA-DR, CD34 and CD45. We assessed the frequency of MSC by limiting dilution assay. RESULTS: The frequency of MSC in BM was significantly higher than in liver, lung, and spleen (p<0.05). On primary non-expanded cells from fetal liver, lung and spleen the number of cells positive for mesenchymal markers was significantly higher within the CD34 positive population than within the CD34 negative population. The phenotype of the culture-expanded MSC was similar for all fetal tissues, i.e. CD90, CD105, CD166, SH3, SH4 and HLA-ABC positive and CD34, CD45 and HLA-DR negative. Culture-expanded cells from all tissues were able to differentiate along adipogenic and osteogenic pathways. However, adipogenic differentiation was less in MSC derived from spleen, and osteogenic differentiation was reduced in liver-derived MSC (p<0.05). INTERPRETATION AND CONCLUSIONS: Our results indicate that culture-expanded MSC derived from second-trimester fetal tissues, although phenotypically similar, exhibit heterogeneity in differentiating potential. We speculate that these differences may be relevant for the clinical application of MSC.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Immunophenotyping , Liver/cytology , Lung/cytology , Mesoderm/cytology , Multipotent Stem Cells/cytology , Spleen/cytology , Abortion, Legal , Adipocytes/chemistry , Adipocytes/classification , Adipocytes/cytology , Adipocytes/metabolism , Antigens, CD34/analysis , Antigens, CD34/immunology , Cells, Cultured , Female , Fetus/cytology , Granulocytes/chemistry , Granulocytes/cytology , Granulocytes/metabolism , Humans , Immunophenotyping/methods , Liver/embryology , Lung/embryology , Lymphocytes/chemistry , Lymphocytes/cytology , Lymphocytes/metabolism , Mesoderm/classification , Monocytes/chemistry , Monocytes/cytology , Monocytes/metabolism , Multipotent Stem Cells/classification , Osteocytes/chemistry , Osteocytes/classification , Osteocytes/cytology , Osteocytes/metabolism , Pregnancy , Pregnancy Trimester, Second , Spleen/embryology
18.
Ann N Y Acad Sci ; 996: 235-44, 2003 May.
Article in English | MEDLINE | ID: mdl-12799302

ABSTRACT

Techniques have recently beome available to isolate and grow mesenchymal progenitors and to manipulate their growth under defined in vitro culture conditions. As a result mesenchymal stem cells can be rapidly expanded to numbers that are required for clinical application. This has allowed the clinical testing of culture-expanded MSCs in the context of hematopoietic stem cell transplantation. In this paper we discuss the role of MSCs in hematopoietic engraftment after transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Mesoderm/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Cell- and Tissue-Based Therapy , Hematopoietic Stem Cell Transplantation/trends , Humans , Mesoderm/immunology
19.
Exp Hematol ; 30(8): 870-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12160838

ABSTRACT

OBJECTIVE: Mesenchymal stem cells (MSC) have been implicated as playing an important role in hematopoietic stem cell engraftment. We identified and characterized a new population of MSC derived from human fetal lung. In cotransplantation experiments, we examined the homing of MSC as well as the effect on engraftment of human umbilical cord blood (UCB)-derived CD34(+) cells in NOD/SCID mice. MATERIALS AND METHODS: Culture-expanded fetal lung-derived CD34(+) cells were characterized by immune phenotyping and cultured under conditions promoting differentiation to osteoblasts or adipocytes. Irradiated (3.5 Gy) NOD/SCID mice (n = 51) were transplanted intravenously with 0.03 to 1.0 x 10(6) UCB CD34(+) cells in the presence or absence of 1 x 10(6) culture-expanded fetal lung-derived MSC, irradiated CD34(-) cells, B cells, or with cultured MSC only. RESULTS: Culture-expanded fetal lung CD34(+) cells were identified as MSC based on phenotype (CD105(+), SH3(+), SH4(+), CD160(+)) and their multilineage potential. Cotransplantation of low doses of UCB CD34(+) cells and MSC resulted in a three-fold to four-fold increase in bone marrow engraftment after 6 weeks, whereas no such effect was observed after cotransplantation of irradiated CD34(-) or B cells. Homing experiments indicated the presence of MSC in the lung, but not in the bone marrow, of NOD/SCID mice. CONCLUSIONS: We identified a population of MSC derived from human fetal lung. Upon cotransplantation, MSC, but not irradiated CD34(-) or B cells, promote engraftment of UCB CD34(+) cells in bone marrow, spleen, and blood by mechanisms that may not require homing of MSC to the bone marrow.


Subject(s)
Fetal Blood/cytology , Fetal Tissue Transplantation , Graft Survival , Hematopoietic Stem Cell Transplantation , Mesoderm/cytology , Stem Cell Transplantation , Adipocytes/cytology , Animals , B-Lymphocytes/radiation effects , Bone Marrow/pathology , Cell Differentiation , Cell Lineage , Cell Movement , Cytokines/metabolism , Hematopoietic Stem Cells/radiation effects , Humans , Lung/cytology , Lung/embryology , Mice , Mice, Inbred NOD , Mice, SCID , Organ Specificity , Osteoblasts/cytology , Radiation Chimera , Stem Cells/metabolism , Stem Cells/physiology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...