Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Haematologica ; 106(2): 483-494, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32001531

RESUMEN

Therapeutic strategies that target leukemic stem cells (LSCs) provide potential advantages in the treatment of chronic myeloid leukemia (CML). Here, we show that selective blockade of plasminogen activator inhibitor-1 (PAI-1) enhances the susceptibility of CML-LSCs to tyrosine kinase inhibitor (TKI), which facilitates the eradication of CML-LSCs and leads to sustained remission of the disease. We demonstrated for the first time that TGF-ß-PAI-1 axis was selectively augmented in CML-LSCs in the bone marrow (BM), whereby protecting CML-LSCs from TKI treatment. Furthermore, the combined administration of TKI plus a PAI-1 inhibitor, in a mouse model of CML, significantly enhanced the eradication of CML cells in the BM and prolonged the survival of CML mice. The combined therapy of imatinib and a PAI-1 inhibitor prevented the recurrence of CML-like disease in serially transplanted recipients, indicating the elimination of CML-LSCs. Interestingly, PAI-1 inhibitor treatment augmented membrane-type matrix metalloprotease-1 (MT1-MMP)-dependent motility of CML-LSCs, and the anti-CML effect of PAI-1 inhibitor was extinguished by the neutralizing antibody for MT1-MMP, underlining the mechanistic importance of MT1-MMP. Our findings provide evidence of, and a rationale for, a novel therapeutic tactic, based on the blockade of PAI-1 activity, for CML patients.


Asunto(s)
Proteínas de Fusión bcr-abl , Leucemia Mielógena Crónica BCR-ABL Positiva , Animales , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Ratones , Células Madre Neoplásicas , Inhibidor 1 de Activador Plasminogénico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Serpina E2
2.
Biochem Biophys Res Commun ; 516(2): 500-505, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31230745

RESUMEN

Upon hematopoietic stem cell transplantation (HSCT), the availability of recipients' niches in the bone marrow (BM) is one of the factors that influence donor HSC engraftment and hematopoietic reconstitution. Therefore, myeloablative conditioning, such as irradiation and/or chemotherapy, which creates empty niches in the recipients' BM, is required for the success of HSCT. However, the conventional myeloablation causes extensive damages to the patients' BM, which results in the treatment-induced severe complications and even mortality. Thus, alternative and mild conditioning could fulfill the need for safer HSCT-based therapies for hematological and nonhematological disorders. Recently, we have demonstrated that pharmacological inhibition of plasminogen activator inhibitor-1 (PAI-1) activity increases cellular motility and cause detachment of HSCs from the niches. In this study, we performed HSCT using a PAI-1 inhibitor without any myeloablative conditioning. Donor HSCs were transplanted to recipient mice that were pretreated with saline or a PAI-1 inhibitor. Saline pretreated nonmyeloablative recipients showed no engraftment. In contrast, donor cell engraftment was detected in the PAI-1 inhibitor pretreated recipients. Multilineage differentiation, including lymphoid and myeloid cells, was observed in the PAI-1 inhibitor pretreated recipients. Donor-derived cells that exhibited multilineage reconstitution as well as the existence of stem/progenitor cells were detected in the secondary recipients, confirming the maintenance of donor HSCs in the BM of PAI-1 inhibitor pretreated primary recipients. The results indicate that the PAI-1 blockade vacates functional niches in the recipients' BM, which allows the engraftment of long-term multilineage HSCs without myeloablative conditioning.


Asunto(s)
Médula Ósea/metabolismo , Trasplante de Células Madre Hematopoyéticas , Inhibidor 1 de Activador Plasminogénico/metabolismo , Nicho de Células Madre , Acondicionamiento Pretrasplante , Animales , Linaje de la Célula , Células Madre Hematopoyéticas/metabolismo , Ratones
3.
Blood ; 130(21): 2283-2294, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-28821477

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) reside in the supportive stromal niche in bone marrow (BM); when needed, however, they are rapidly mobilized into the circulation, suggesting that HSPCs are intrinsically highly motile but usually stay in the niche. We questioned what determines the motility of HSPCs. Here, we show that transforming growth factor (TGF)-ß-induced intracellular plasminogen activator inhibitor (PAI)-1 activation is responsible for keeping HSPCs in the BM niche. We found that the expression of PAI-1, a downstream target of TGF-ß signaling, was selectively augmented in niche-residing HSPCs. Functional inhibition of the TGF-ß-PAI-1 signal increased MT1-MMP-dependent cellular motility, causing a detachment of HSPCs from the TGF-ß-expressing niche cells, such as megakaryocytes. Furthermore, consistently high motility in PAI-1-deficient HSPCs was demonstrated by both a transwell migration assay and reciprocal transplantation experiments, indicating that intracellular, not extracellular, PAI-1 suppresses the motility of HSPCs, thereby causing them to stay in the niche. Mechanistically, intracellular PAI-1 inhibited the proteolytic activity of proprotein convertase Furin, diminishing MT1-MMP activity. This reduced expression of MT1-MMP in turn affected the expression levels of several adhesion/deadhesion molecules for determination of HSPC localization, such as CD44, VLA-4, and CXCR4, which then promoted the retention of HSPCs in the niche. Our findings open up a new field for the study of intracellular proteolysis as a regulatory mechanism of stem cell fate, which has the potential to improve clinical HSPC mobilization and transplantation protocols.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Espacio Intracelular/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Nicho de Células Madre , Factor de Crecimiento Transformador beta/metabolismo , Animales , Médula Ósea/metabolismo , Movimiento Celular , Espacio Extracelular/metabolismo , Furina/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Transducción de Señal
4.
J Cell Physiol ; 232(9): 2569-2580, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27735989

RESUMEN

Adult bone mass is maintained through a balance of the activities of osteoblasts and osteoclasts. Although Notch signaling has been shown to maintain bone homeostasis by controlling the commitment, differentiation, and function of cells in both the osteoblast and osteoclast lineages, the precise mechanisms by which Notch performs such diverse and complex roles in bone physiology remain unclear. By using a transgenic approach that modified the expression of delta-like 1 (DLL1) or Jagged1 (JAG1) in an osteoblast-specific manner, we investigated the ligand-specific effects of Notch signaling in bone homeostasis. This study demonstrated for the first time that the proper regulation of DLL1 expression, but not JAG1 expression, in osteoblasts is essential for the maintenance of bone remodeling. DLL1-induced Notch signaling was responsible for the expansion of the bone-forming cell pool by promoting the proliferation of committed but immature osteoblasts. However, DLL1-Notch signaling inhibited further differentiation of the expanded osteoblasts to become fully matured functional osteoblasts, thereby substantially decreasing bone formation. Osteoblast-specific expression of DLL1 did not alter the intrinsic differentiation ability of cells of the osteoclast lineage. However, maturational arrest of osteoblasts caused by the DLL1 transgene impaired the maturation and function of osteoclasts due to a failed osteoblast-osteoclast coupling, resulting in severe suppression of bone metabolic turnover. Taken together, DLL1-mediated Notch signaling is critical for proper bone remodeling as it regulates the differentiation and function of both osteoblasts and osteoclasts. Our study elucidates the importance of ligand-specific activation of Notch signaling in the maintenance of bone homeostasis. J. Cell. Physiol. 232: 2569-2580, 2017. © 2016 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals Inc.


Asunto(s)
Remodelación Ósea , Fémur/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Osteoblastos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/metabolismo , Tibia/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Proteínas de Unión al Calcio , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Femenino , Fémur/embriología , Fémur/crecimiento & desarrollo , Genotipo , Edad Gestacional , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Osteoclastos/metabolismo , Osteogénesis , Fenotipo , Tibia/embriología , Tibia/crecimiento & desarrollo
5.
Blood ; 118(11): 2941-50, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21734240

RESUMEN

Stem cells of highly regenerative organs including blood are susceptible to endogenous DNA damage caused by both intrinsic and extrinsic stress. Response mechanisms to such stress equipped in hematopoietic stem cells (HSCs) are crucial in sustaining hematopoietic homeostasis but remain largely unknown. In this study, we demonstrate that serial transplantation of human HSCs into immunodeficient mice triggers replication stress that induces incremental elevation of intracellular reactive oxygen species (ROS) levels and the accumulation of persistent DNA damage within the human HSCs. This accumulation of DNA damage is also detected in HSCs of clinical HSC transplant patients and elderly individuals. A forced increase of intracellular levels of ROS by treatment with a glutathione synthetase inhibitor aggravates the extent of DNA damage, resulting in the functional impairment of HSCs in vivo. The oxidative DNA damage activates the expression of cell-cycle inhibitors in a HSC specific manner, leading to the premature senescence among HSCs, and ultimately to the loss of stem cell function. Importantly, treatment with an antioxidant can antagonize the oxidative DNA damage and eventual HSC dysfunction. The study reveals that ROS play a causative role for DNA damage and the regulation of ROS have a major influence on human HSC aging.


Asunto(s)
Proliferación Celular , Daño del ADN/fisiología , Células Madre Hematopoyéticas/fisiología , Estrés Oxidativo/fisiología , Animales , Supervivencia Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo
6.
Am J Pathol ; 177(2): 547-54, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20558580

RESUMEN

Mesenchymal stem cells (MSCs) have emerged as a new therapeutic modality for reconstituting the hematopoietic microenvironment by improving engraftment in stem cell transplantation. However, the availability of conventional bone marrow (BM)-derived MSCs (BMSCs) is limited. Recent studies showed that a large number of MSCs can be easily isolated from fat tissue (adipose tissue-derived MSCs [ADSCs]). In this study, we extensively evaluated the hematopoiesis-supporting properties of ADSCs, which are largely unknown. In vitro coculture and progenitor assays showed that ADSCs generated significantly more granulocytes and progenitor cells from human hematopoietic stem cells (HSCs) than BMSCs. We found that ADSCs express the chemokine CXCL12, a critical regulator of hematopoiesis, at levels that are three fold higher than those with BMSCs. The addition of a CXCL12 receptor antagonist resulted in a lower yield of granulocytes from ADSC layers, whereas the addition of recombinant CXCL12 to BMSC cocultures promoted the growth of granulocytes. In vivo cell homing assays showed that ADSCs facilitated the homing of mouse HSCs to the BM better than BMSCs. ADSCs injected into the BM cavity of fatally irradiated mice reconstituted hematopoiesis more promptly than BMSCs and subsequently rescued mice that had received a low number of HSCs. Secondary transplantation experiments showed that ADSCs exerted favorable effects on long-term HSCs. These results suggest that ADSCs can be a promising therapeutic alternative to BMSCs.


Asunto(s)
Tejido Adiposo/citología , Células de la Médula Ósea/fisiología , Hematopoyesis/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Línea Celular , Quimiocina CXCL12/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL
7.
Haematologica ; 96(4): 543-51, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21193418

RESUMEN

BACKGROUND: To understand how myelodysplastic syndrome cells evolve from normal stem cells and gain competitive advantages over normal hematopoiesis, we established a murine xenograft model harboring bone marrow cells from patients with myelodysplastic syndromes or acute myeloid leukemia with myelodysplasia-related changes. DESIGN AND METHODS: Bone marrow CD34(+) cells obtained from patients were injected, with or without human mesenchymal stem cells, into the bone marrow of non-obese diabetic/severe combined immunodeficient/IL2Rγ(null) hosts. Engraftment and differentiation of cells derived from the patients were investigated by flow cytometry and immunohistochemical analysis. RESULTS: Co-injection of patients' cells and human mesenchymal stem cells led to successful engraftment of patient-derived cells that maintained the immunophenotypes and genomic abnormalities of the original patients. Myelodysplastic syndrome-originated clones differentiated into mature neutrophils, megakaryocytes, and erythroblasts. Two of the samples derived from patients with acute myeloid leukemia with myelodysplasia-related changes were able to sustain neoplastic growth into the next generation while these cells had limited differentiation ability in the murine host. The hematopoiesis of mice engrafted with patients' cells was significantly suppressed even when human cells accounted for less than 1% of total marrow mononuclear cells. Histological studies revealed invasion of the endosteal surface by patient-derived CD34(+) cells and disruption of extracellular matrix architecture, which probably caused inhibition of murine hematopoiesis. CONCLUSIONS: We established murine models of human myelodysplastic syndromes using cells obtained from patients: the presence of neoplastic cells was associated with the suppression of normal host hematopoiesis. The efficiency of engraftment was related to the presence of an abnormality in chromosome 7.


Asunto(s)
Modelos Animales de Enfermedad , Síndromes Mielodisplásicos/patología , Trasplante Heterólogo , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Noqueados , Ratones SCID , Síndromes Mielodisplásicos/metabolismo
8.
Stem Cells ; 26(12): 3228-36, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18787204

RESUMEN

Hematopoiesis is a dynamic and strictly regulated process orchestrated by self-renewing hematopoietic stem cells (HSCs) and the supporting microenvironment. However, the exact mechanisms by which individual human HSCs sustain hematopoietic homeostasis remain to be clarified. To understand how the long-term repopulating cell (LTRC) activity of individual human HSCs and the hematopoietic hierarchy are maintained in the bone marrow (BM) microenvironment, we traced the repopulating dynamics of individual human HSC clones using viral integration site analysis. Our study presents several lines of evidence regarding the in vivo dynamics of human hematopoiesis. First, human LTRCs existed in a rare population of CD34(+)CD38(-) cells that localized to the stem cell niches and maintained their stem cell activities while being in a quiescent state. Second, clonally distinct LTRCs controlled hematopoietic homeostasis and created a stem cell pool hierarchy by asymmetric self-renewal division that produced lineage-restricted short-term repopulating cells and long-lasting LTRCs. Third, we demonstrated that quiescent LTRC clones expanded remarkably to reconstitute the hematopoiesis of the secondary recipient. Finally, we further demonstrated that human mesenchymal stem cells differentiated into key components of the niche and maintained LTRC activity by closely interacting with quiescent human LTRCs, resulting in more LTRCs. Taken together, this study provides a novel insight into repopulation dynamics, turnover, hierarchical structure, and the cell cycle status of human HSCs in the recipient BM microenvironment.


Asunto(s)
Células de la Médula Ósea/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , ADP-Ribosil Ciclasa 1/biosíntesis , Antígenos CD34/biosíntesis , Médula Ósea/metabolismo , Técnicas de Cultivo de Célula/métodos , Ciclo Celular , Sangre Fetal/metabolismo , Homeostasis , Humanos , Lentivirus/metabolismo , Células Madre Mesenquimatosas/citología , Trasplante de Células Madre , Factores de Tiempo
9.
Exp Hematol ; 35(12): 1872-83, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17923243

RESUMEN

OBJECTIVE: Hematopoietic stem cells (HSCs) consist of heterogenous subpopulations, one of which is CD34(-) HSCs. Recent development of successful engraftment by intra-bone marrow transplantation revealed severe combined immunodeficiency (scid) mouse-repopulating cell (SRC) activity in human CD34(-) cord blood (CB) cells. On the other hand, CD34(-) cells from bone marrow (BM) cells remain relatively undefined. Here, we investigated pre-SRC populations in human BM CD34(-) cells and the effect of the niche-related factor, angiopoietin-1, on them. METHODS: Two populations in BM CD34(-) cells (namely M cells and S cells) were purified by flow cytometry. Then, they were cocultured with six growth factors on the hematopoietic-supportive mouse BM stromal cell line, HESS-5 or AHESS-5 that were engineered to produce human angiopoietin-1, because we detected Tie2 expression on M cells and S cells. Cultured cells were assessed for their in vitro and in vivo hematopietic activities. RESULTS: After 7 days in coculture, AHESS-5 was stronger more effective than HESS-5 in converting M and S cells to CD34(+) cells (M cells: 67.4% vs 17.5%, n =6, p < 0.001) (S cells: 42.3% vs 2.3%, n = 6, p < 0.001). Furthermore, both M and S cells were able to engraft in immunodeficient mice after they were cocultured on AHESS-5. CONCLUSIONS: Results suggest that angiopoietin-1 supports SRC activities in human CD34(-) BM cells, as murine studies demonstrated. Furthermore, identification of previously undetected subpopulations of BM CD34(-) HSCs unveils heterogenous components in the stem cell pool.


Asunto(s)
Angiopoyetina 1/fisiología , Antígenos CD34/inmunología , Células de la Médula Ósea/citología , Hematopoyesis , Western Blotting , Células de la Médula Ósea/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica
10.
PLoS One ; 12(6): e0179239, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28617827

RESUMEN

Immunodeficient mice transplanted with human peripheral blood mononuclear cells (PBMCs) are promising tools to evaluate human immune responses to vaccines. However, these mice usually develop severe graft-versus-host disease (GVHD), which makes estimation of antigen-specific IgG production after antigen immunization difficult. To evaluate antigen-specific IgG responses in PBMC-transplanted immunodeficient mice, we developed a novel NOD/Shi-scid-IL2rγnull (NOG) mouse strain that systemically expresses the human IL-4 gene (NOG-hIL-4-Tg). After human PBMC transplantation, GVHD symptoms were significantly suppressed in NOG-hIL-4-Tg compared to conventional NOG mice. In kinetic analyses of human leukocytes, long-term engraftment of human T cells has been observed in peripheral blood of NOG-hIL-4-Tg, followed by dominant CD4+ T rather than CD8+ T cell proliferation. Furthermore, these CD4+ T cells shifted to type 2 helper (Th2) cells, resulting in long-term suppression of GVHD. Most of the human B cells detected in the transplanted mice had a plasmablast phenotype. Vaccination with HER2 multiple antigen peptide (CH401MAP) or keyhole limpet hemocyanin (KLH) successfully induced antigen-specific IgG production in PBMC-transplanted NOG-hIL-4-Tg. The HLA haplotype of donor PBMCs might not be relevant to the antibody secretion ability after immunization. These results suggest that the human PBMC-transplanted NOG-hIL-4-Tg mouse is an effective tool to evaluate the production of antigen-specific IgG antibodies.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Formación de Anticuerpos , Proteínas Portadoras/inmunología , Inmunización , Inmunoglobulina G/inmunología , Interleucina-4/inmunología , Péptidos/farmacología , Receptor ErbB-2/farmacología , Animales , Anticuerpos Antineoplásicos/genética , Linfocitos B/inmunología , Linfocitos B/trasplante , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Proteínas Portadoras/genética , Xenoinjertos , Humanos , Inmunoglobulina G/genética , Interleucina-4/genética , Ratones , Ratones SCID , Ratones Transgénicos , Péptidos/inmunología , Receptor ErbB-2/inmunología , Células Th2/inmunología , Células Th2/trasplante
11.
Exp Hematol ; 31(12): 1323-30, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14662341

RESUMEN

OBJECTIVE: Recent studies have shown that bone marrow (BM) contains cells capable of differentiating into myocytes in vivo. However, addition of demethylation drugs has been necessary to induce myocyte differentiation from BM cells in vitro, and precise mechanisms of BM cells' conversion to myocytes and the origin of those cells have not been established. We investigated the expression of myogenic markers during differentiation and maturation of myocytes from BM-derived multipotent adult progenitor cells (MAPC) under physiological culture condition. MATERIALS AND METHODS: Frozen BM samples from 21 healthy donors were used as a source of MAPC. To induce myocyte differentiation MAPC was cultured in the presence of 5% FCS, VEGF, bFGF, and IGF-1, and the expressions of myocyte markers were examined at various time points. We also investigated engraftment and differentiation of MAPC-derived myocytes in vivo. RESULTS: Frozen BM-derived MAPC, cultured under the physiological myogenic condition, demonstrated spatial expression patterns of several myocyte markers similar to that of authentic myocyte differentiation. When injected into murine muscles, MAPC treated with the myogenic condition engrafted and differentiated into myocyte marker-positive cells and myotubes in vivo. CONCLUSION: For the first time, we were able to induce myocyte formation from BM cells under the physiological condition in vitro and demonstrated that treating cells with this condition prior to intramuscular injection increased efficiency of engraftment and differentiation in vivo.


Asunto(s)
Células de la Médula Ósea , Células Madre Multipotentes/citología , Células Musculares/citología , Células Musculares/trasplante , Adolescente , Adulto , Animales , Antígenos CD/análisis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Linaje de la Célula , Trasplante de Células , Niño , Supervivencia de Injerto , Humanos , Ratones , Persona de Mediana Edad , Fibras Musculares Esqueléticas/citología , Trasplante Heterólogo
12.
PLoS One ; 9(11): e111082, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25369030

RESUMEN

Recent advances in cancer biology have revealed that many malignancies possess a hierarchal system, and leukemic stem cells (LSC) or leukemia-initiating cells (LIC) appear to be obligatory for disease progression. Acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia characterized by the formation of a PML-RARα fusion protein, leads to the accumulation of abnormal promyelocytes. In order to understand the precise mechanisms involved in human APL leukemogenesis, we established a humanized in vivo APL model involving retroviral transduction of PML-RARA into CD34(+) hematopoietic cells from human cord blood and transplantation of these cells into immunodeficient mice. The leukemia well recapitulated human APL, consisting of leukemic cells with abundant azurophilic abnormal granules in the cytoplasm, which expressed CD13, CD33 and CD117, but not HLA-DR and CD34, were clustered in the same category as human APL samples in the gene expression analysis, and demonstrated sensitivity to ATRA. As seen in human APL, the induced APL cells showed a low transplantation efficiency in the secondary recipients, which was also exhibited in the transplantations that were carried out using the sorted CD34- fraction. In order to analyze the mechanisms underlying APL initiation and development, fractionated human cord blood was transduced with PML-RARA. Common myeloid progenitors (CMP) from CD34(+)/CD38(+) cells developed APL. These findings demonstrate that CMP are a target fraction for PML-RARA in APL, whereas the resultant CD34(-) APL cells may share the ability to maintain the tumor.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Animales , Antígenos CD34/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Humanos , Leucemia Promielocítica Aguda/mortalidad , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos NOD , Proteínas de Fusión Oncogénica/genética , Tasa de Supervivencia , Trasplante Heterólogo
14.
Stem Cells ; 25(5): 1213-21, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17218403

RESUMEN

Mesenchymal stem cells (MSC), a distinct type of adult stem cell, are easy to isolate, culture, and manipulate in ex vivo culture. These cells have great plasticity and potential for therapeutic application, but their properties are poorly understood because of their low frequency and the lack of knowledge on cell surface markers and their location of origin. The present study was designed to address the undefined lineage relationship of hematopoietic and mesenchymal stem cells. Genetically marked, highly purified hematopoietic stem cells (HSCs) were transplanted into wild-type animals and, after bone marrow repopulation, the progeny were rigorously investigated for differentiation potential into mesenchymal tissues by analyzing in vitro differentiation into mesenchymal tissues. None/very little of the hematopoietic cells contributed to colony-forming units fibroblast activity and mesenchymal cell differentiation; however, unfractionated bone marrow cells resulted in extensive replacement of not only hematopoietic cells but also mesenchymal cells, including MSCs. As a result, we concluded that purified HSCs have no significant potency to differentiate into mesenchymal lineage. The data strongly suggest that hematopoietic cells and mesenchymal lineage cells are derived from individual lineage-specific stem cells. In addition, we succeeded in visualizing mesenchymal lineage cells using in vivo microimaging and immunohistochemistry. Flow cytometric analysis revealed CD140b (PDGFRbeta) could be a specific marker for mesenchymal lineage cells. The results may reinforce the urgent need for a more comprehensive view of the mesenchymal stem cell identity and characteristics. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula , Células Madre/citología , Animales , Biomarcadores/metabolismo , Adhesión Celular , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Sistema Hematopoyético/citología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Blood ; 107(5): 1878-87, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16282345

RESUMEN

Hematopoiesis is maintained by specific interactions between both hematopoietic and nonhematopoietic cells. Whereas hematopoietic stem cells (HSCs) have been extensively studied both in vitro and in vivo, little is known about the in vivo characteristics of stem cells of the nonhematopoietic component, known as mesenchymal stem cells (MSCs). Here we have visualized and characterized human MSCs in vivo following intramedullary transplantation of enhanced green fluorescent protein-marked human MSCs (eGFP-MSCs) into the bone marrow (BM) of nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. Between 4 to 10 weeks after transplantation, eGFP-MSCs that engrafted in murine BM integrated into the hematopoietic microenvironment (HME) of the host mouse. They differentiated into pericytes, myofibroblasts, BM stromal cells, osteocytes in bone, bone-lining osteoblasts, and endothelial cells, which constituted the functional components of the BM HME. The presence of human MSCs in murine BM resulted in an increase in functionally and phenotypically primitive human hematopoietic cells. Human MSC-derived cells that reconstituted the HME appeared to contribute to the maintenance of human hematopoiesis by actively interacting with primitive human hematopoietic cells.


Asunto(s)
Diferenciación Celular/fisiología , Hematopoyesis/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID
16.
Blood ; 108(7): 2446-54, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16757689

RESUMEN

To elucidate the in vivo kinetics of human hematopoietic stem cells (HSCs), CD34+CD38- cells were infected with lentivirus vector and transplanted into immunodeficient mice. We analyzed the multilineage differentiation and self-renewal abilities of individual thymus-repopulating clones in primary recipients, and their descending clones in paired secondary recipients, by tracing lentivirus gene integration sites in each lymphomyeloid progeny using a linear amplification-mediated polymerase chain reaction (PCR) strategy. Our clonal analysis revealed that a single human thymus-repopulating cell had the ability to produce lymphoid and myeloid lineage cells in the primary recipient and each secondary recipient, indicating that individual human HSCs expand clonally by self-renewal division. Furthermore, we found that the proportion of HSC clones present in the CD34+ cell population decreased as HSCs replicated during extensive repopulation and also as the differentiation capacity of the HSC clones became limited. This indicates the restriction of the ability of individual HSCs despite the expansion of total HSC population. We also demonstrated that the extensive self-renewal potential was confined in the relatively small proportion of HSC clones. We conclude that our clonal tracking studies clearly demonstrated that heterogeneity in the self-renewal capacity of HSC clones underlies the differences in clonal longevity in the CD34+ stem cell pool.


Asunto(s)
Células Madre Hematopoyéticas/citología , Timo/citología , ADP-Ribosil Ciclasa 1/biosíntesis , Animales , Antígenos CD34/biosíntesis , Diferenciación Celular , Linaje de la Célula , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Humanos , Cinética , Lentivirus/genética , Ratones , Ratones SCID , Modelos Biológicos , Timo/metabolismo
17.
Blood ; 101(8): 2905-13, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12411299

RESUMEN

To measure the ability of human hematopoietic stem cells (HSCs), the SCID-repopulating cell (SRC) assay has been widely used. Conventionally, human HSCs are transplanted into a nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse via a tail vein. However, those cells must go through various obstacles until they reach the mouse marrow environment, which could explain the generally low homing efficiency in this system. Thus, the capability of HSCs may not be studied accurately by this intravenous transplantation method. In our attempt to reveal actual SRC potential, ie, self-renewal and multilineage differentiation in recipient bone marrow, we introduced cells into mouse marrow directly (intrabone marrow [iBM]) to minimize the effect of factors that may interfere with the homing of HSCs and compared the results obtained by intravenous and iBM methods. When cord blood CD34(+)CD38(-) cells were transplanted in NOD/SCID mice by iBM, a 15-fold higher frequency of SRC, 1 in 44 CD34(+)CD38(-) cells, was achieved compared with 1 in 660 by the intravenous method. Furthermore, the iBM transplant showed high levels of engraftment in the secondary transplantation. Pretreatment of CD34(+) cells with antibodies that block either very late antigen 4 (VLA-4) or VLA-5 reduced engraftment partially, whereas blockage of both molecules resulted in complete inhibition of engraftment, which suggests that VLA-4 and VLA-5 are involved in different processes in engraftment or have complementary roles. Our results indicate that the iBM injection strategy is a more sensitive and direct way to measure the capability of human SRCs and is useful to investigate the interaction of HSCs and marrow environment in vivo.


Asunto(s)
Médula Ósea , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Células Madre Hematopoyéticas/citología , Trasplante Heterólogo/métodos , Animales , Linaje de la Célula , Movimiento Celular , Femenino , Citometría de Flujo , Genes Reporteros , Supervivencia de Injerto , Proteínas Fluorescentes Verdes , Humanos , Recién Nacido , Inyecciones , Inyecciones Intravenosas , Integrina alfa4beta1/fisiología , Integrina alfa5beta1/fisiología , Proteínas Luminiscentes/análisis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Especificidad de Órganos , Receptores CXCR4/fisiología , Proteínas Recombinantes de Fusión/análisis , Cola (estructura animal)/irrigación sanguínea , Quimera por Trasplante
18.
Blood ; 104(12): 3581-7, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15297308

RESUMEN

Bone marrow (BM) cells are reported to contribute to the process of regeneration following myocardial infarction. However, the responsible BM cells have not been fully identified. Here, we used 2 independent clonal studies to determine the origin of bone marrow (BM)-derived cardiomyocytes. First, we transplanted single CD34(-) c-kit(+)Sca-1(+) lineage(-) side population (CD34(-)KSL-SP) cells or whole BM cells from mice ubiquitously expressing enhanced green fluorescent protein (EGFP) into lethally irradiated mice, induced myocardial infarction (MI), and treated the animals with granulocyte colony-stimulating factor (G-CSF) to mobilize stem cells to the damaged myocardium. At 8 weeks after MI, from 100 specimens we counted only 3 EGFP(+) actinin(+) cells in myocardium of CD34(-) KSL-SP cells in mice that received transplants, but more than 5000 EGFP(+) actinin(+) cells in whole BM cell in mice that received transplants, suggesting that most of EGFP(+) actinin(+) cells were derived from nonhematopoietic BM cells. Next, clonally purified nonhematopoietic mesenchymal stem cells (MSCs), cardiomyogenic (CMG) cells, that expressed EGFP in the cardiomyocyte-specific manner were transplanted directly into BM of lethally irradiated mice, MI was induced, and they were treated with G-CSF. EGFP(+) actinin(+) cells were observed in the ischemic myocardium, indicating that CMG cells had been mobilized and differentiated into cardiomyocytes. Together, these results suggest that the origin of the vast majority of BM-derived cardiomyocytes is MSCs.


Asunto(s)
Quimiotaxis , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Animales , Trasplante de Médula Ósea , Diferenciación Celular , Linaje de la Célula , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Regeneración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA