Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
Biol Aujourdhui ; 210(1): 9-17, 2016.
Artigo em Francês | MEDLINE | ID: mdl-27286576

RESUMO

Human adult pluripotent stem cells, stem cells of embryonic origin and induced pluripotent stem cells (iPS) provide cellular sources for new promising regenerative medicine approaches. Because these cells can be patient-specific, they allow considering a personalized medicine appropriate to the diagnosis of each. The generation of cultured red blood cells (cRBC) derived from stem cells is emblematic of personalized medicine. Indeed, these cells have the advantage of being selected according to a blood phenotype of interest and they may provide treatments to patients in situation of impossible transfusion (alloimmunized patients, rare phenotypes). Essential progresses have established proof of concept for this approach, still a concept some years ago. From adult stem cells, all steps of upstream research were successfully achieved, including the demonstration of the feasibility of injection into human. This leads us to believe that Red Blood Cells generated in vitro from stem cells will be the future players of blood transfusion. However, although theoretically ideal, these stem cells raise many biological challenges to overcome, although some tracks are identified.


Assuntos
Eritrócitos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Medicina Regenerativa/tendências , Adulto , Células-Tronco Adultas/fisiologia , Técnicas de Cultura de Células/métodos , Células Cultivadas , Humanos , Medicina de Precisão/métodos , Medicina de Precisão/tendências
3.
Stem Cells ; 33(8): 2431-41, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25850942

RESUMO

While enucleation is a critical step in the terminal differentiation of human red blood cells, the molecular mechanisms underlying this unique process remain unclear. To investigate erythroblast enucleation, we studied the erythroid differentiation of human embryonic stem cells (hESCs), which provide a unique model for deeper understanding of the development and differentiation of multiple cell types. First, using a two-step protocol, we demonstrated that terminal erythroid differentiation from hESCs is directly dependent on the age of the embryoid bodies. Second, by choosing hESCs in two extreme conditions of erythroid culture, we obtained an original differentiation model which allows one to study the mechanisms underlying the enucleation of erythroid cells by analyzing the gene and miRNA (miR) expression profiles of cells from these two culture conditions. Third, using an integrated analysis of mRNA and miR expression profiles, we identified five miRs potentially involved in erythroblast enucleation. Finally, by selective knockdown of these five miRs we found miR-30a to be a regulator of erythroblast enucleation in hESCs.


Assuntos
Diferenciação Celular , Eritroblastos/metabolismo , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas/metabolismo , MicroRNAs/biossíntese , RNA Mensageiro/biossíntese , Células Cultivadas , Eritroblastos/citologia , Células-Tronco Embrionárias Humanas/citologia , Humanos
4.
PLoS One ; 8(5): e62303, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658722

RESUMO

A role for caspase activation in erythroid differentiation has been established, yet its precise mode of action remains elusive. A drawback of all previous investigations on caspase activation in ex vivo erythroid differentiation is the lack of an in vitro model producing full enucleation of erythroid cells. Using a culture system which renders nearly 100% enucleated red cells from human CD34(+) cells, we investigated the role of active caspase-3 in erythropoiesis. Profound effects of caspase-3 inhibition were found on erythroid cell growth and differentiation when inhibitors were added to CD34(+) cells at the start of the culture and showed dose-response to the concentration of inhibitor employed. Enucleation was only reduced as a function of the reduced maturity of the culture and the increased cell death of mature cells while the majority of cells retained their ability to extrude their nuclei. Cell cycle analysis after caspase-3 inhibition showed caspase-3 to play a critical role in cell proliferation and highlighted a novel function of this protease in erythroid differentiation, i.e. its contribution to cell cycle regulation at the mitotic phase. While the effect of caspase-3 inhibitor treatment on CD34(+) derived cells was not specific to the erythroid lineage, showing a similar reduction of cell expansion in myeloid cultures, the mechanism of action in both lineages appeared to be distinct with a strong induction of apoptosis causing the decreased yield of myeloid cells. Using a series of colony-forming assays we were able to pinpoint the stage at which cells were most sensitive to caspase-3 inhibition and found activated caspase-3 to play a signalling role in erythroid differentiation by targeting mature BFU-E and CFU-E but not early BFU-E.


Assuntos
Caspase 3/metabolismo , Diferenciação Celular , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/enzimologia , Transdução de Sinais , Apoptose/efeitos dos fármacos , Inibidores de Caspase/farmacologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Células Precursoras Eritroides/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
5.
Am J Hematol ; 88(5): 343-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23417980

RESUMO

Lentiviral modification combined with ex vivo erythroid differentiation was used to stably inhibit RhAG expression, a critical component of the Rh(rhesus) membrane complex defective in the Rh(null) syndrome. The cultured red cells generated recapitulate the major alterations of native Rh(null) cells regarding antigen expression, membrane deformability, and gas transport function, providing the proof of principle for their use as model of Rh(null) syndrome and to investigate Rh complex biogenesis in human primary erythroid cells. Using this model, we were able to reveal for the first time that RhAG extinction alone is sufficient to explain ICAM-4 and CD47 loss observed on native Rh(null) RBCs. Together with the effects of RhAG forced expression in Rh(null) progenitors, this strongly strengthens the hypothesis that RhAG is critical to Rh complex formation. The strategy is also promising for diagnosis purpose in order to overcome the supply from rare blood donors and is applicable to other erythroid defects and rare phenotypes, providing models to dissect membrane biogenesis of multicomplex proteins in erythroid cells, with potential clinical applications in transfusion medicine.


Assuntos
Proteínas Sanguíneas/metabolismo , Antígeno CD47/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Eritroides/metabolismo , Doenças Genéticas Inatas/metabolismo , Glicoproteínas de Membrana/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Anemia Hemolítica Congênita/metabolismo , Anemia Hemolítica Congênita/patologia , Anemia Hipoplástica Congênita/metabolismo , Anemia Hipoplástica Congênita/patologia , Proteínas Sanguíneas/antagonistas & inibidores , Proteínas Sanguíneas/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Células Eritroides/patologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Sangue Fetal , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Doenças Genéticas Inatas/sangue , Doenças Genéticas Inatas/patologia , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Gravidez , Interferência de RNA , RNA Interferente Pequeno , Reticulócitos/metabolismo , Reticulócitos/patologia , Sistema do Grupo Sanguíneo Rh-Hr/sangue
6.
Haematologica ; 97(12): 1795-803, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22733021

RESUMO

BACKGROUND: Human induced pluripotent stem cells offer perspectives for cell therapy and research models for diseases. We applied this approach to the normal and pathological erythroid differentiation model by establishing induced pluripotent stem cells from normal and homozygous sickle cell disease donors. DESIGN AND METHODS: We addressed the question as to whether these cells can reach complete erythroid terminal maturation notably with a complete switch from fetal to adult hemoglobin. Sickle cell disease induced pluripotent stem cells were differentiated in vitro into red blood cells and characterized for their terminal maturation in terms of hemoglobin content, oxygen transport capacity, deformability, sickling and adherence. Nucleated erythroblast populations generated from normal and pathological induced pluripotent stem cells were then injected into non-obese diabetic severe combined immunodeficiency mice to follow the in vivo hemoglobin maturation. RESULTS: We observed that in vitro erythroid differentiation results in predominance of fetal hemoglobin which rescues the functionality of red blood cells in the pathological model of sickle cell disease. We observed, in vivo, the switch from fetal to adult hemoglobin after infusion of nucleated erythroid precursors derived from either normal or pathological induced pluripotent stem cells into mice. CONCLUSIONS: These results demonstrate that human induced pluripotent stem cells: i) can achieve complete terminal erythroid maturation, in vitro in terms of nucleus expulsion and in vivo in terms of hemoglobin maturation; and ii) open the way to generation of functionally corrected red blood cells from sickle cell disease induced pluripotent stem cells, without any genetic modification or drug treatment.


Assuntos
Anemia Falciforme/patologia , Diferenciação Celular , Eritrócitos/patologia , Eritropoese/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Adulto , Líquido Amniótico/química , Anemia Falciforme/metabolismo , Animais , Adesão Celular , Células Cultivadas , Eritrócitos/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Hemoglobinas/metabolismo , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Oxigênio/metabolismo
7.
Stem Cells Int ; 2011: 405429, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21961016

RESUMO

We previously described the large-scale production of RBCs from hematopoietic stem cells (HSCs) of diverse sources. Our present efforts are focused to produce RBCs thanks to an unlimited source of stem cells. Human embryonic stem (ES) cells or induced pluripotent stem cell (iPS) are the natural candidates. Even if the proof of RBCs production from these sources has been done, their amplification ability is to date not sufficient for a transfusion application. In this work, our protocol of RBC production was applied to HSC isolated from fetal liver (FL) as an intermediate source between embryonic and adult stem cells. We studied the erythroid potential of FL-derived CD34(+) cells. In this in vitro model, maturation that is enucleation reaches a lower level compared to adult sources as observed for embryonic or iP, but, interestingly, they (i) displayed a dramatic in vitro expansion (100-fold more when compared to CB CD34(+)) and (ii) 100% cloning efficiency in hematopoietic progenitor assays after 3 days of erythroid induction, as compared to 10-15% cloning efficiency for adult CD34(+) cells. This work supports the idea that FL remains a model of study and is not a candidate for ex vivo RBCS production for blood transfusion as a direct source of stem cells but could be helpful to understand and enhance proliferation abilities for primitive cells such as ES cells or iPS.

8.
Blood ; 118(19): 5071-9, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21885599

RESUMO

In vitro RBC production from stem cells could represent an alternative to classic transfusion products. Until now the clinical feasibility of this concept has not been demonstrated. We addressed the question of the capacity of cultured RBCs (cRBCs) to survive in humans. By using a culture protocol permitting erythroid differentiation from peripheral CD34(+) HSC, we generated a homogeneous population of cRBC functional in terms of their deformability, enzyme content, capacity of their hemoglobin to fix/release oxygen, and expression of blood group antigens. We then demonstrated in the nonobese diabetes/severe combined immunodeficiency mouse that cRBC encountered in vivo the conditions necessary for their complete maturation. These data provided the rationale for injecting into one human a homogeneous sample of 10(10) cRBCs generated under good manufacturing practice conditions and labeled with (51)Cr. The level of these cells in the circulation 26 days after injection was between 41% and 63%, which compares favorably with the reported half-life of 28 ± 2 days for native RBCs. Their survival in vivo testifies globally to their quality and functionality. These data establish the proof of principle for transfusion of in vitro-generated RBCs and path the way toward new developments in transfusion medicine. This study is registered at http://www.clinicaltrials.gov as NCT0929266.


Assuntos
Transfusão de Eritrócitos/métodos , Animais , Antígenos CD34/sangue , Antígenos de Grupos Sanguíneos/sangue , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Envelhecimento Eritrocítico , Deformação Eritrocítica , Eritrócitos/citologia , Eritrócitos/imunologia , Eritrócitos/metabolismo , Eritropoese , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Hemoglobinas/metabolismo , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante Heterólogo
9.
Curr Opin Hematol ; 18(4): 249-53, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21519239

RESUMO

PURPOSE OF REVIEW: In the context of chronic blood supply difficulties, generating cultured red blood cells (cRBCs) in vitro after amplification of stem cells makes sense. This review will focus on the recent findings about the generation of erythroid cells from induced pluripotent stem (iPS) cells and deals with the hurdles and next developments that will occur. RECENT FINDINGS: The most proliferative source of stem cells for generating cRBCs is the cord blood, but this source is limited in terms of hematopoietic stem cells and dependent on donations. Pluripotent stem cells are thus the best candidates and potential sources of cRBCs. Critical advances have led towards the in-vitro production of functional RBCs from iPS cells in the last few years. SUMMARY: Because iPS cells can proliferate indefinitely and can be selected for a phenotype of interest, they are potential candidates to organize complementary sources of RBCs for transfusion. Proof of concept of generating cRBCs from iPS cells has been performed, but the procedures need to be optimized to lead to clinical application in blood transfusion. Several crucial points remain to be resolved. Notably these include the choice of the initial cell type to generate iPS cells, the method of reprogramming, that is, to ensure the safety of iPS cells as clinical grade, the optimization of erythrocyte differentiation, and the definition of good manufacturing practice (GMP) conditions for industrial production.


Assuntos
Eritrócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Transfusão de Sangue/métodos , Humanos , Camundongos
10.
Haematologica ; 95(10): 1651-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20494935

RESUMO

BACKGROUND: Ex vivo manufacture of red blood cells from stem cells is a potential means to ensure an adequate and safe supply of blood cell products. Advances in somatic cell reprogramming of human induced pluripotent stem cells have opened the door to generating specific cells for cell therapy. Human induced pluripotent stem cells represent a potentially unlimited source of stem cells for erythroid generation for transfusion medicine. DESIGN AND METHODS: We characterized the erythroid differentiation and maturation of human induced pluripotent stem cell lines obtained from human fetal (IMR90) and adult fibroblasts (FD-136) compared to those of a human embryonic stem cell line (H1). Our protocol comprises two steps: (i) differentiation of human induced pluripotent stem cells by formation of embryoid bodies with indispensable conditioning in the presence of cytokines and human plasma to obtain early erythroid commitment, and (ii) differentiation/maturation to the stage of cultured red blood cells in the presence of cytokines. The protocol dispenses with major constraints such as an obligatory passage through a hematopoietic progenitor, co-culture on a cellular stroma and use of proteins of animal origin. RESULTS: We report for the first time the complete differentiation of human induced pluripotent stem cells into definitive erythrocytes capable of maturation up to enucleated red blood cells containing fetal hemoglobin in a functional tetrameric form. CONCLUSIONS: Red blood cells generated from human induced pluripotent stem cells pave the way for future development of allogeneic transfusion products. This could be done by banking a very limited number of red cell phenotype combinations enabling the safe transfusion of a great number of immunized patients.


Assuntos
Eritrócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular , Citocinas/farmacologia , Transfusão de Eritrócitos , Humanos
11.
Haematologica ; 95(3): 398-405, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19815832

RESUMO

BACKGROUND: Anemia is a characteristic of myelodysplastic syndromes, such as the rare 5q- syndrome, but its mechanism remains unclear. In particular, data are lacking on the terminal phase of differentiation of erythroid cells (enucleation) in myelodysplastic syndromes. DESIGN AND METHODS: We used a previously published culture model to generate mature red blood cells in vitro from human hematopoietic progenitor cells in order to study the pathophysiology of the 5q- syndrome. Our model enables analysis of cell proliferation and differentiation at a single cell level and determination of the enucleation capacity of erythroid precursors. RESULTS: The erythroid commitment of 5q(del) clones was not altered and their terminal differentiation capacity was preserved since they achieved final erythroid maturation (enucleation stage). The drop in red blood cell production was secondary to the decrease in the erythroid progenitor cell pool and to impaired proliferative capacity. RPS14 gene haploinsufficiency was related to defective erythroid proliferation but not to differentiation capacity. CONCLUSIONS: The 5q- syndrome should be considered a quantitative rather than qualitative bone marrow defect. This observation might open the way to new therapeutic concepts.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/genética , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Eritrócitos/citologia , Células Precursoras Eritroides/fisiologia , Síndromes Mielodisplásicas/genética , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Eritropoese , Feminino , Humanos , Masculino , Síndromes Mielodisplásicas/patologia , Proteínas Ribossômicas/genética
12.
Cell Immunol ; 253(1-2): 16-22, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18639869

RESUMO

The interactions between mesenchymal stem cells (MSCs) and immune system are currently being explored. Leukemia inhibitory factor (LIF) is linked to regulatory transplantation tolerance. Our aim was to study the expression of LIF on human MSCs at both gene and protein level in mixed lymphocyte reaction (MSC/MLR), and its implication in MSC immunosuppressive effect. There was a 7-fold increase (611pg/ml) in LIF in MSC/MLR as compared to MSCs alone. Using LIF neutralizing antibody, a significant restoration of up to 91% of CD3+ lymphocyte proliferation in MSC/MLR was observed (p=0.021). LIF was implicated in the generation of regulatory lymphocytes, as demonstrated by decrease of Foxp3+ regulatory cells after using LIF neutralizing antibody in MSC/MLR (p=0.06) by flow cytometry. A positive correlation between LIF and human leukocyte antigen (HLA-G) gene expression by MSCs was found (R(2)=0.74). Our findings provide evidence supporting the immunomodulatory effect of MSCs.


Assuntos
Tolerância Imunológica/imunologia , Terapia de Imunossupressão , Fator Inibidor de Leucemia/imunologia , Células-Tronco Mesenquimais/imunologia , Anticorpos/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Separação Celular , Células Cultivadas , Fatores de Transcrição Forkhead/imunologia , Antígenos HLA/imunologia , Antígenos HLA-G , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células-Tronco Mesenquimais/citologia
13.
Leuk Lymphoma ; 48(10): 2032-41, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17917971

RESUMO

Osteolytic bone lesions are common in patients with multiple myeloma (MM), a clonal plasma cell disorder, and result from increased osteoclastic bone resorption and decreased osteoblastic bone formation. Because mesenchymal stem cells (MSCs) are committed towards cells of the osteoblast lineage, we compared the in vitro characteristics of MSCs from the bone marrow of 18 MM patients (MM-MSCs) and eight normal donors (ND-MSCs). MM-MSCs displayed deficient growth that could be explained in part by the reduced expression of several growth factor receptors on the surface of MM-MSCs compared with ND-MSCs. Receptor downregulation was observed on RT-PCR analysis. A major finding was an approximately fivefold higher expression of osteoblast inhibitor DKK1 at transcript and protein levels in MM-MSCs than ND-MSCs. These data suggest that defective osteoblast function in patients with advanced MM may be related not only to factors released by tumor myeloma cells but also to MSC abnormalities.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Idoso , Osso e Ossos/metabolismo , Diferenciação Celular , Feminino , Humanos , Imunofenotipagem , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/metabolismo , Masculino , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteólise , Sindecana-1/biossíntese
14.
Transplantation ; 84(2): 231-7, 2007 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-17667815

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) possess unique immunomodulatory properties. They are able to suppress allogenic T-cell response and modify maturation of antigen-presenting cells. Their role in the treatment of severe graft versus host disease has been reported. The underlying molecular mechanisms of immunosuppression are currently being investigated. Histocompatibility locus antigen (HLA)-G is a nonclassical major histocompatibility complex class I antigen with strong immune-inhibitory properties. METHODS: We studied the role of HLA-G on MSC-induced immunosuppression. The expression of HLA-G on human MSCs cultured alone and in mixed lymphocytes reaction (MSC/MLR) was analyzed. RESULTS: We found that HLA-G can be detected on MSCs by real-time reverse-phase polymerase chain reaction, immunofluorescence, flow cytometry (52.4+/-3.6%), and enzyme-linked immunosorbent assay in the supernatant (38.7+/-5.2 ng/mL). HLA-G protein expression is constitutive and the level is not modified upon stimulation by allogenic lymphocytes in MSC/MLR. The functional role of HLA-G protein expressed by MSCs was analyzed using the 87G anti-HLA-G blocking antibody in a MSC/MLR. We found that blocking HLA-G molecule significantly raised lymphocyte proliferation in MSC/MLR (35.5%, P=0.01). CONCLUSION: Our findings provide evidences supporting involvement of HLA-G in the immunosuppressive properties of MSCs. These results emphasize the potential application of MSCs as a relevant therapeutic candidate in transplantation.


Assuntos
Expressão Gênica , Antígenos HLA/genética , Antígenos HLA/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Terapia de Imunossupressão/métodos , Células-Tronco Mesenquimais/imunologia , RNA Mensageiro/genética , Células da Medula Óssea/citologia , Proliferação de Células , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Antígenos HLA/metabolismo , Antígenos HLA-G , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia
15.
Gene Expr ; 13(4-5): 217-26, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17605296

RESUMO

Mesenchymal stem cells (MSC) inhibit the response of allogeneic T lymphocytes in culture. Because the mechanisms of this effect may differ according to the existence of cell contact, we investigated the differences in gene expression of inhibitory molecules during MSC-T lymphocyte coculture when cell contact does and does not occur. Human MSC and T lymphocytes were cultured together in standard and transwell cultures. MSC gene expression was analyzed by semiquantitative real-time RT-PCR. MSC elicited a high dose-dependent inhibition of T lymphocytes in cultures with cell contact, but inhibition occurred even without cell contact. In both cases, we observed significant upregulation of IDO, LIF, and HLA-G, along with downregulation of HGF and SDF1. In cultures with cell contact, IL-10 and TGF-beta transcripts were expressed in a significantly higher level than in cultures without this contact. Furthermore, in the latter, the increased inhibition of T-cell proliferation was positively correlated with IDO gene expression and negatively correlated with SDF1 gene expression. MSC appear to induce T-cell tolerance by two distinct mechanisms. The first of these, which does not require cell contact, induces expression of the tolerogenic genes IDO, LIF, and HLA-G. The second mechanism, which is contact dependent, modulates IL-10 and TGF-beta gene expression. These two mechanisms probably play separate roles in MSC-induced tolerance in allogeneic hematopoietic stem cell transplantation.


Assuntos
Comunicação Celular/fisiologia , Regulação da Expressão Gênica , Interleucina-10/metabolismo , Células-Tronco Mesenquimais/fisiologia , Linfócitos T/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Moléculas de Adesão Celular/metabolismo , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Técnicas de Cocultura , Humanos , Terapia de Imunossupressão , Interleucina-10/genética , Células-Tronco Mesenquimais/citologia , Linfócitos T/citologia , Fator de Crescimento Transformador beta/genética
16.
Leuk Lymphoma ; 47(7): 1340-7, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16923566

RESUMO

This study evaluated the feasibility of using dendritic cells (DCs) to generate, ex vivo, anti-tumor cytotoxic T lymphocytes (CTL) in patients with stage III multiple myeloma (MM). Nucleated cells from eight patients who had received chemotherapy (three of whom had undergone autologous hemopoeitic stem cell transplantation) were collected by apheresis. Their monocytes were enriched using counter-current centrifugation, differentiated into DCs which were further co-cultured with autologous CD8 lymphocytes to induce CTL. The DCs were pulsed either with the idiotypic paraprotein (regarded as a tumor-specific antigen) or with autologous MM cell lysate before co-culture. Specific T-cell responses were measured in IFNgamma enzyme-linked immunospot and chromium release assays of autologous plasmocyte targets. A slight increase in IFNgamma secretion by T-cells was observed for two patients (DCs pulsed with idiotypic paraprotein for one, MM cell lysate for the other). No or weak specific lysis of plasmocyte targets was observed in the chromium release assays. In conclusion, the T-cell response to pulsed DCs was very weak or absent. There are clinical and technical reasons that could explain, in part, this lack of response.


Assuntos
Transplante de Células/métodos , Células Dendríticas/citologia , Idiótipos de Imunoglobulinas , Mieloma Múltiplo/terapia , Linfócitos T/metabolismo , Adulto , Idoso , Remoção de Componentes Sanguíneos , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Cromo/metabolismo , Técnicas de Cocultura , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Mieloma Múltiplo/imunologia , Fator de Necrose Tumoral alfa/biossíntese
17.
Stem Cells ; 24(4): 1020-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16339642

RESUMO

Mesenchymal stem cells (MSCs) have been shown to migrate to various tissues. There is little information on the fate and potential therapeutic efficacy of the reinfusion of MSCs following total body irradiation (TBI). We addressed this question using human MSC (hMSCs) infused to nonobese diabetic/ severe combined immunodeficient (NOD/SCID) mice submitted to TBI. Further, we tested the impact of additional local irradiation (ALI) superimposed to TBI, as a model of accidental irradiation. NOD/SCID mice were transplanted with hM-SCs. Group 1 was not irradiated before receiving hMSC infusion. Group 2 received only TBI at a dose of 3.5 Gy, group 3 received local irradiation to the abdomen at a dose of 4.5 Gy in addition to TBI, and group 4 received local irradiation to the leg at 26.5 Gy in addition to TBI. Fifteen days after irradiation, quantitative and spatial distribution of the hMSCs were studied. Histological analysis of mouse tissues confirmed the presence of radio-induced lesions in the irradiated fields. Following their infusion into nonirradiated animals, hMSCs homed at a very low level to various tissues (lung, bone marrow, and muscles) and no significant engraftment was found in other organs. TBI induced an increase of engraftment levels of hMSCs in the brain, heart, bone marrow, and muscles. Abdominal irradiation (AI) as compared with leg irradiation (LI) increased hMSC engraftment in the exposed area (the gut, liver, and spleen). Hind LI as compared with AI increased hMSC engraftment in the exposed area (skin, quadriceps, and muscles). An increase of hMSC engraftment in organs outside the fields of the ALI was also observed. Conversely, following LI, hMSC engraftment was increased in the brain as compared with AI. This study shows that engraftment of hMSCs in NOD/ SCID mice with significantly increased in response to tissue injuries following TBI with or without ALI. ALI induced an increase of the level of engraftment at sites outside the local irradiation field, thus suggesting a distant (abscopal) effect of radiation damage. This work supports the use of MSCs to repair damaged normal tissues following accidental irradiation and possibly in patients submitted to radiotherapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos da radiação , Animais , Movimento Celular/efeitos da radiação , Expressão Gênica , Globinas/genética , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/efeitos da radiação , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Especificidade de Órgãos , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/terapia , Transplante Heterólogo , Irradiação Corporal Total , Microglobulina beta-2/metabolismo
18.
Zhonghua Yi Xue Za Zhi ; 85(39): 2780-4, 2005 Oct 19.
Artigo em Chinês | MEDLINE | ID: mdl-16324321

RESUMO

OBJECTIVE: To evaluate whether mesenchymal stem cells (MSCs) obtained from human proximal femurs possess immunosuppressive effect so as to look for ideal bank of MSCs for clinical prophylaxis and treatment of graft versus host disease (GVHD). METHODS: Human marrows were collected from the proximal femurs of patients undergoing hip replacement to isolate MSCs. The puncture materials obtained from the iliac bone marrow of 12 healthy donors were used as controls. Peripheral blood lymphocytes (PBLs) were obtained from the peripheral blood of healthy persons. 1 x 10(5) PBLs were mixed with allogeneic PBLs radiated by 60 cobalt and put into the wells of a 96-well plate. MSCs of the concentrations of 1 x 10(5), 3 x 10(4), 1 x 10(4), and 3 x 10(3), were added into the culture fluid of the mixed PBLs to be co-cultivated for 5 days. 1 microCi/well [(3)H] TdR was added in the last 18 hours. The cells were collected and the counts per minute (cpm) was detected. 3 x 10(4) and 1 x 10(4) MSCs were put into the wells. When the MSCs adhered to the wall, a membrane with micropores was inserted value of 1 x 10(5) PBLs and radiated allo-PBLs were put onto the top of which. Five days after cultivation 1 microCi/well [(3)H] TdR was added and the cpm was tested. MSCs were cultured in RPMI-1640 culture fluid and then contacted MLR constructed by 1 x 10(5) PBLs and 1 x 10(5) allo-PBL directly or via Transwell membrane with micropores. Five days after the supernatant was collected. ELISA was used to detect the content of TGF-beta(1). 0.1 microg/ml, 1 microg/ml, or 10 microg/ml anti-human TGF-beta1 antibody was added to the co-cultivation system. Five days after [(3)H] TdR was added so as to test the value cpm. RESULTS: 3 x 10(3) - 1 x 10(5) MSCs from proximal femurs inhibited the PBLs proliferation to 62 +/- 18% - 28 +/- 12% of maximal response, however, not significantly different from that observed in MSCs collected from bone marrow of healthy donors via iliac crest aspiration (58 +/- 12% - 27 +/- 6%, P > 0.05). When these cells were separated physically from MLR system via a membrane with micropores 0.2 microm in diameter, 3 x 10(4) and 1 x 10(4) cells still markedly suppressed the PBLs proliferation to 36 +/- 8% and 53 +/- 13% of maximal response. ELISA showed that TGF-beta1 was measured in the supernatants of MSCs, MSCs + MLR and MSCs + MLR in Transwell system, without significant differences among these experimental conditions. Furthermore, the presence of increasing amounts of neutralizing anti- TGF-beta1 antibody did not reverse the inhibitory effect. CONCLUSION: MSCs obtained from the proximal femurs possess immunosuppressive activity. A soluble factor/factors was/were involved in such effect, however, TGF-beta1 was not a candidate.


Assuntos
Fêmur/citologia , Tolerância Imunológica , Linfócitos/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Adulto , Idoso , Células da Medula Óssea/citologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
19.
Blood ; 103(9): 3313-9, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14715641

RESUMO

The Stro-1 antigen potentially defines a mesenchymal stem cell (MSC) progenitor subset. We here report on the role of human ex vivo-expanded selected Stro-1(+) or Stro-1(-) MSC subsets on the engraftment of human CD34(+) cord blood cells in the nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mouse model. The data show that cotransplantation of expanded Stro-1(-) cells with CD34(+) cells resulted in a significant increase of human CD45, CD34, CD19, and CD11b cells detected in blood or in bone marrow (BM) and spleen as compared with the infusion of CD34(+) cells alone. Infusion into mice of expanded Stro-1(+) and Stro-1(-) cells (without CD34(+) cells) showed that the numbers of Stro-1(+)-derived (as assessed by DNA analysis of human beta-globin with quantitative polymerase chain reaction [PCR]) were higher than Stro-1(-)-derived cells in spleen, muscles, BM, and kidneys, while more Stro-1(-)-derived than Stro-1(+)-derived cells were found in lungs. The transduction of expanded Stro-1(+) cells with an enhanced green fluorescent protein (eGFP) gene did not modify their cytokine release and their homing in NOD/SCID mouse tissues. The difference between the hematopoietic support and the homing capabilities of expanded Stro-1(+) and Stro-1(-) cells may be of importance for clinical therapeutic applications: Stro-1(+) cells may rather be used for gene delivery in tissues while Stro-1(-) cells may rather be used to support hematopoietic engraftment.


Assuntos
Movimento Celular , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Proteínas Nucleares , Animais , Células da Medula Óssea , Células Cultivadas , Sobrevivência de Enxerto , Hematopoese , Humanos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos SCID , Modelos Animais , Especificidade de Órgãos , Baço/citologia , Transplante Heterólogo
20.
J Gene Med ; 5(12): 1028-38, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14661178

RESUMO

BACKGROUND: Recent studies have suggested that ex vivo expansion of autologous hematopoietic cells could be a therapy of choice for the treatment of bone marrow failure. We investigated the potential of a combined infusion of autologous ex vivo expanded hematopoietic cells with mesenchymal (MSCs) for the treatment of multi-organ failure syndrome following irradiation in a non-human primate model. METHODS: Hematopoietic cells and MSCs were expanded from bone marrow aspirates. MSCs were transduced with the gene encoding for the green fluorescent protein (e-GFP), in order to track them following infusion. Twelve animals were studied. Nine animals received total-body irradiation at 8 Gy from a neutron/gamma source thus resulting in heterogeneous exposure; three animals were sham-irradiated. The animals were treated with expanded hematopoietic stem cells and MSCs, expanded hematopoietic stem cells alone, or MSCs alone. Unmanipulated bone marrow cell transplants were used as controls. RESULTS: Depending on the neutron/gamma ratio, an acute radiation sickness of varying severity but of similar nature resulted. GFP-labeled cells were found in the injured muscle, skin, bone marrow and gut of the treated animals via PCR up to 82 days post-infusion. CONCLUSIONS: This is the first evidence of expanded MSCs homing in numerous tissues following a severe multi-organ injury in primates. Localization of the transduced MSCs correlated to the severity and geometry of irradiation. A repair process was observed in various tissues. The plasticity potential of the MSCs and their contribution to the repair process in vivo remains to be studied.


Assuntos
Movimento Celular , Transplante de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Mesenquimais , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/terapia , Lesões por Radiação/complicações , Lesões por Radiação/terapia , Animais , Transplante de Medula Óssea , Terapia Combinada , Marcadores Genéticos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Macaca fascicularis , Masculino , Transdução Genética , Resultado do Tratamento , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...