Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 128(25): 2949-2959, 2016 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-27733356

RESUMO

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.


Assuntos
Medula Óssea/patologia , Leucemia Mieloide Aguda/patologia , Nicho de Células-Tronco , Alicerces Teciduais/química , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Autorrenovação Celular , Separação Celular , Células Clonais , Feminino , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Leucemia Mieloide Aguda/genética , Células-Tronco Mesenquimais/citologia , Camundongos , Fenótipo , Células Estromais/patologia
3.
Haematologica ; 101(5): 616-25, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26858358

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Imunoterapia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão , Linfócitos T/imunologia , Linfócitos T/metabolismo , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/imunologia , Animais , Citocinas/biossíntese , Citotoxicidade Imunológica , Modelos Animais de Doenças , Citometria de Fluxo , Expressão Gênica , Técnicas de Transferência de Genes , Genes Transgênicos Suicidas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante , Transdução Genética , Carga Tumoral/genética , Carga Tumoral/imunologia
4.
Clin Cancer Res ; 21(12): 2802-10, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25398450

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/antagonistas & inibidores , ADP-Ribosil Ciclase 1/metabolismo , Bortezomib/farmacologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Talidomida/análogos & derivados , Adulto , Idoso , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Bortezomib/administração & dosagem , Linhagem Celular Tumoral , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Humanos , Imunoterapia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Lenalidomida , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/terapia , Talidomida/administração & dosagem , Talidomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-22653974

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/citologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Nicho de Células-Tronco/imunologia , Quimeras de Transplante/imunologia , Microambiente Tumoral/imunologia , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Ossículos da Orelha/citologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Camundongos , Camundongos Mutantes , Transplante de Neoplasias , Osteólise/imunologia , Alicerces Teciduais , Transplante Heterólogo
6.
Br J Haematol ; 148(3): 428-40, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19863541

RESUMO

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.


Assuntos
Feto/citologia , Células-Tronco Mesenquimais/fisiologia , Actinas/metabolismo , Tecido Adiposo/citologia , Adulto , Medula Óssea/embriologia , Células da Medula Óssea/fisiologia , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Quimiotaxia/fisiologia , Humanos , Integrinas/metabolismo , Pulmão/citologia , Pulmão/embriologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Paxilina/metabolismo , Fosforilação , Células Estromais/citologia , Células Estromais/metabolismo , Células Estromais/fisiologia
7.
Pediatr Cardiol ; 30(5): 699-709, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19184178

RESUMO

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.


Assuntos
Coração/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Medula Óssea , Cardiopatias/terapia , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Transplante de Células-Tronco
8.
EuroIntervention ; 4(1): 133-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-19112790

RESUMO

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.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Separação Celular/normas , Leucócitos Mononucleares/citologia , Infarto do Miocárdio/cirurgia , Esterno/citologia , Anticoagulantes/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Soluções Tampão , Procedimentos Cirúrgicos Cardíacos , Movimento Celular , Centrifugação/métodos , Protocolos Clínicos , Ensaios Clínicos como Assunto , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Heparina/farmacologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Albumina Sérica/farmacologia , Transplante de Células-Tronco
9.
J Immunol ; 180(7): 5141-8, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18354240

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Endoteliais/imunologia , Receptores de Lipopolissacarídeos/imunologia , Células-Tronco/imunologia , Complexo CD3/imunologia , Comunicação Celular , Proliferação de Células , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Monócitos/citologia , Monócitos/imunologia , Neovascularização Fisiológica/genética , Fenótipo , Receptores de Antígenos de Linfócitos T/imunologia
10.
Exp Hematol ; 33(10): 1249-56, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16219548

RESUMO

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.


Assuntos
Doadores de Sangue , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sobrevivência de Enxerto/fisiologia , Hematopoese/fisiologia , Recuperação de Função Fisiológica/fisiologia , Adulto , Animais , Antígenos CD34/metabolismo , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/citologia , Sangue Fetal/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Teste de Histocompatibilidade , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante Heterólogo , Transplante Homólogo
11.
Exp Hematol ; 31(10): 881-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14550803

RESUMO

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.


Assuntos
Antígenos CD34/análise , Transplante de Medula Óssea , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Pulmão/citologia , Mesoderma/citologia , Animais , Movimento Celular , Humanos , Imunofenotipagem , Fígado/citologia , Camundongos , Camundongos Nus , Camundongos SCID
12.
Haematologica ; 88(8): 845-52, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12935972

RESUMO

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.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Imunofenotipagem , Fígado/citologia , Pulmão/citologia , Mesoderma/citologia , Células-Tronco Multipotentes/citologia , Baço/citologia , Aborto Legal , Adipócitos/química , Adipócitos/classificação , Adipócitos/citologia , Adipócitos/metabolismo , Antígenos CD34/análise , Antígenos CD34/imunologia , Células Cultivadas , Feminino , Feto/citologia , Granulócitos/química , Granulócitos/citologia , Granulócitos/metabolismo , Humanos , Imunofenotipagem/métodos , Fígado/embriologia , Pulmão/embriologia , Linfócitos/química , Linfócitos/citologia , Linfócitos/metabolismo , Mesoderma/classificação , Monócitos/química , Monócitos/citologia , Monócitos/metabolismo , Células-Tronco Multipotentes/classificação , Osteócitos/química , Osteócitos/classificação , Osteócitos/citologia , Osteócitos/metabolismo , Gravidez , Segundo Trimestre da Gravidez , Baço/embriologia
14.
Ann N Y Acad Sci ; 996: 235-44, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12799302

RESUMO

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.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Mesoderma/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Terapia Baseada em Transplante de Células e Tecidos , Transplante de Células-Tronco Hematopoéticas/tendências , Humanos , Mesoderma/imunologia
15.
Exp Hematol ; 30(8): 870-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12160838

RESUMO

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.


Assuntos
Sangue Fetal/citologia , Transplante de Tecido Fetal , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Mesoderma/citologia , Transplante de Células-Tronco , Adipócitos/citologia , Animais , Linfócitos B/efeitos da radiação , Medula Óssea/patologia , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Citocinas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Pulmão/citologia , Pulmão/embriologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Especificidade de Órgãos , Osteoblastos/citologia , Quimera por Radiação , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...