Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Transfus Med Hemother ; 43(4): 247-254, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27721700

RESUMEN

Hematopoietic stem cells (HSCs) are the best characterized adult stem cells and the only stem cell type in routine clinical use. The concept of stem cell transplantation laid the foundations for the development of novel cell therapies within, and even outside, the hematopoietic system. Here, we report on the history of hematopoietic cell transplantation (HCT) and of HSC isolation, we briefly summarize the capabilities of HSCs to reconstitute the entire hemato/lymphoid cell system, and we assess current indications for HCT. We aim to draw the lines between areas where HCT has been firmly established, areas where HCT can in the future be expected to be of clinical benefit using their regenerative functions, and areas where doubts persist. We further review clinical trials for diverse approaches that are based on HCT. Finally, we highlight the advent of genome editing in HSCs and critically view the use of HSCs in non-hematopoietic tissue regeneration.

2.
Stem Cells Dev ; 25(12): 922-33, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27100571

RESUMEN

Polycomb proteins such as the B lymphoma Mo-MLV insertion region 1 homolog (BMI1) are essential chromatin factors for the self-renewal and differentiation of embryonic and adult stem cells. BMI1 also plays a critical role in osteogenesis as Bmi1-deficient mice display a skeletal phenotype caused by the exhaustion of the mesenchymal stem cell pool. In this study, we have studied the role of BMI1 in the osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs). BMI1 protein, but not RNA levels, increases during in vitro osteogenic differentiation of hASCs. Overexpression of BMI1 leads to an osteogenic priming of hASCs under nondifferentiating conditions and enhanced osteogenesis upon differentiation, along with increased BMP2 and WNT11 expressions. Conversely, knockdown of BMI1 expression reduces osteogenic differentiation. Furthermore, our studies indicate that during osteogenic differentiation of hASCs, BMI1 is a downstream target of GSK3 signaling. BMI1, therefore, acts as a pro-osteogenic differentiation factor in hASCs and hence it is a promising target for active modulation of hASC-derived osteogenesis.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Complejo Represivo Polycomb 1/metabolismo , Fosfatasa Alcalina/metabolismo , Matriz Ósea/metabolismo , Calcificación Fisiológica , Células Cultivadas , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Transducción de Señal
3.
Sci Rep ; 5: 12319, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-26198814

RESUMEN

Cord blood hematopoietic stem cells (CB-HSCs) are an outstanding source for transplantation approaches. However, the amount of cells per donor is limited and culture expansion of CB-HSCs is accompanied by a loss of engraftment potential. In order to analyze the molecular mechanisms leading to this impaired potential we profiled global and local epigenotypes during the expansion of human CB hematopoietic stem and progenitor cells (HPSCs). Human CB-derived CD34+ cells were cultured in serum-free medium together with SCF, TPO, FGF, with or without Igfbp2 and Angptl5 (STF/STFIA cocktails). As compared to the STF cocktail, the STFIA cocktail maintains in vivo repopulation capacity of cultured CD34+ cells. Upon expansion, CD34+ cells genome-wide remodel their epigenotype and depending on the cytokine cocktail, cells show different H3K4me3 and H3K27me3 levels. Expanding cells without Igfbp2 and Angptl5 leads to higher global H3K27me3 levels. ChIPseq analyses reveal a cytokine cocktail-dependent redistribution of H3K27me3 profiles. Inhibition of the PRC2 component EZH2 counteracts the culture-associated loss of NOD scid gamma (NSG) engraftment potential. Collectively, our data reveal chromatin dynamics that underlie the culture-associated loss of engraftment potential. We identify PRC2 component EZH2 as being involved in the loss of engraftment potential during the in vitro expansion of HPSCs.


Asunto(s)
Sangre Fetal/citología , Supervivencia de Injerto/fisiología , Células Madre Hematopoyéticas/citología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/metabolismo , Animales , Antígenos CD34/metabolismo , Células Cultivadas , Cromatina/metabolismo , Femenino , Sangre Fetal/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones SCID , Trasplante Heterólogo/métodos
4.
Mol Med ; 21: 185-96, 2015 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-25811991

RESUMEN

Parent-of-origin imprints have been implicated in the regulation of neural differentiation and brain development. Previously we have shown that, despite the lack of a paternal genome, human parthenogenetic (PG) embryonic stem cells (hESCs) can form proliferating neural stem cells (NSCs) that are capable of differentiation into physiologically functional neurons while maintaining allele-specific expression of imprinted genes. Since biparental ("normal") hESC-derived NSCs (N NSCs) are targeted by immune cells, we characterized the immunogenicity of PG NSCs. Flow cytometry and immunocytochemistry revealed that both N NSCs and PG NSCs exhibited surface expression of human leukocyte antigen (HLA) class I but not HLA-DR molecules. Functional analyses using an in vitro mixed lymphocyte reaction assay resulted in less proliferation of peripheral blood mononuclear cells (PBMC) with PG compared with N NSCs. In addition, natural killer (NK) cells cytolyzed PG less than N NSCs. At a molecular level, expression analyses of immune regulatory factors revealed higher HLA-G levels in PG compared with N NSCs. In line with this finding, MIR152, which represses HLA-G expression, is less transcribed in PG compared with N cells. Blockage of HLA-G receptors ILT2 and KIR2DL4 on natural killer cell leukemia (NKL) cells increased cytolysis of PG NSCs. Together this indicates that PG NSCs have unique immunological properties due to elevated HLA-G expression.


Asunto(s)
Diferenciación Celular , Citotoxicidad Inmunológica , Células Madre Embrionarias/citología , Expresión Génica , Antígenos HLA-G/genética , Células Asesinas Naturales/inmunología , Células-Madre Neurales/inmunología , Células-Madre Neurales/metabolismo , Apoptosis/genética , Apoptosis/inmunología , Línea Celular , Regulación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Antígenos HLA-G/inmunología , Antígenos HLA-G/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , MicroARNs/genética , Células-Madre Neurales/citología
5.
J Tissue Eng Regen Med ; 7(12): 944-54, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22653714

RESUMEN

Cord blood-derived haematopoietic stem cells (CB-HSCs) are an attractive source for transplantation in haematopoietic disorders. However, the yield of CB-HSCs per graft is limited and often insufficient, particularly for the treatment of adult patients. Here we compare the capacity of three cytokine cocktails to expand CB-CD34(+) cells. Cells were cultured for 5 or 14 days in media supplemented with: (a) SCF, FL, IL-3 and IL-6 (SFLIL3/6); (b) SCF, TPO, FGF-1 and IL-6 (STFIL6); and (c) SCF, TPO, FGF-1, IGFBP2 and Angptl-5 (STFAI). We observed that STFAI-culture expansion sustained the most vigorous cell proliferation, maintenance of CD34(+) phenotype and colony-forming unit counts. In addition, STFAI-cultured cells had a potent ex vivo migration activity. STFAI-expanded cells were able to engraft NSG mice. However, no significant difference in overall engraftment was observed among the expansion cocktails. Assessment of short-term reconstitution using multilineage markers demonstrated that the STFAI cocktail for HSCs expansion greatly improved total cell expansion but may impair short-term lymphoid repopulation.


Asunto(s)
Angiopoyetinas/farmacología , Antígenos CD34/metabolismo , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Linfocitos/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Citocinas/farmacología , Sangre Fetal/metabolismo , Humanos , Antígenos Comunes de Leucocito/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Bazo/citología , Factor de Células Madre/farmacología , Trombopoyetina/farmacología
6.
Tissue Eng Part C Methods ; 19(1): 25-38, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22712684

RESUMEN

Currently, ex vivo expansion of hematopoietic stem cells (HSC) is still insufficient. Traditional approaches for HSC expansion include the use of stromal cultures, growth factors, and/or bioreactors. Biomaterial-based strategies provide new perspectives. We focus on identifying promising two-dimensional (2D) polymer candidates for HSC expansion. After a 7-day culture period with cytokine supplementation, 2D fibrin, poly(D,L-lactic-co-glycolic acid; Resomer® RG503), and Poly(ɛ-caprolactone; PCL) substrates supported expansion of cord blood (CB)-derived CD34⁺ cells ex vivo. Fibrin cultures achieved the highest proliferation rates (>8700-fold increase of total nuclear cells, p<0.001), high total colony-forming units (3.6-fold increase, p<0.001), and highest engraftment in NSG mice (7.69-fold more donor cells compared with tissue culture polysterene, p<0.001). In addition, the presence of multiple human hematopoietic lineages such as myeloid (CD13⁺), erythroid (GypC⁺), and lymphoid (CD20⁺/CD56⁺) in murine transplant recipients confirmed the multilineage engraftment potential of fibrin-based cultures. Filopodia development in fibrin-expanded cells was a further indicator for superior cell adhesion capacities. We propose application of fibrin, Resomer® RG503, and PCL for future strategies of CB-CD34⁺ cell expansion. Suitable polymers for HSC expansion might also be appropriate for future drug discovery applications or for studies aimed to develop hematological therapies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Sangre Fetal/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Polímeros/farmacología , Animales , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula , Supervivencia Celular , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/ultraestructura , Humanos , Inmunohistoquímica , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Especificidad de Órganos/efectos de los fármacos , Ensayo de Tumor de Célula Madre
7.
Cell Med ; 5(1): 29-42, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26858862

RESUMEN

Uniparental zygotes with two paternal (androgenetic, AG) or two maternal genomes (gynogenetic, GG) cannot develop into viable offsprings but form blastocysts from which pluripotent embryonic stem (ES) cells can be derived. For most organs, it is unclear whether uniparental ES cells can give rise to stably expandable somatic stem cells that can repair injured tissues. Even if previous reports indicated that the capacity of AG ES cells to differentiate in vitro into pan-neural progenitor cells (pNPCs) and into cells expressing neural markers is similar to biparental [normal fertilized (N)] ES cells, their potential for functional neurogenesis is not known. Here we show that murine AG pNPCs give rise to neuron-like cells, which then generate sodium-driven action potentials while maintaining fidelity of imprinted gene expression. Neural engraftment after intracerebral transplantation was achieved only by late (22 days) AG and N pNPCs with in vitro low colony-forming cell (CFC) capacity. However, persisting CFC formation seen, in particular, in early (13 or 16 days) differentiation cultures of N and AG pNPCs correlated with a high incidence of trigerm layer teratomas. As AG ES cells display functional neurogenesis and in vivo stability similar to N ES cells, they represent a unique model system to study the roles of paternal and maternal genomes on neural development and on the development of imprinting-associated brain diseases.

8.
Cytotherapy ; 14(5): 570-83, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22300364

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) are promising candidates for innovative cell therapeutic applications. For clinical-scale manufacturing, different supplements have been evaluated as alternatives for the commonly used fetal bovine serum (FBS). We have reported previously that pooled human AB serum (HS) accelerates the proliferation of adipose tissue-derived MSC (ASC) while maintaining key functions of MSC biology such as differentiation, immune suppression and growth factor secretion. ASC expanded in FBS-supplemented culture media undergo replicative aging that is associated with a progressive loss of differentiation capacity but without indications of cellular transformation. The effects of HS media on ASC long-term culture, however, remain poorly characterized. METHODS: Long-term cultures of ASC in FBS and HS media were analyzed with respect to proliferation, marker expression, differentiation and immune suppression. RESULTS: Despite signs of an accelerated proliferation, extended life span and clonogenic capacity of ASC cultivated in HS-supplemented media, HS and FBS cultures revealed no significant differences with respect to differentiation potential and expression of senescence markers. Anchorage-independent growth, which is indicative of tumorigenic properties, was not observed in either culture conditions. Similarly, immune suppressive activities were maintained. Donor variation regarding differentiation potential and marker expression became apparent in this study independent of the culture supplement or culture duration. CONCLUSIONS: We have demonstrated that the use of pooled allogeneic HS maintains the characteristics of ASC even after long-term expansion, further demonstrating that the use of HS is an alternative to FBS.


Asunto(s)
Tejido Adiposo/citología , Técnicas de Cultivo de Célula , Medios de Cultivo , Células Madre Mesenquimatosas/citología , Suero , Tejido Adiposo/metabolismo , Adulto , Animales , Bovinos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/farmacología , Femenino , Fibroblastos/citología , Humanos , Persona de Mediana Edad
9.
Exp Hematol ; 39(6): 617-28, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21356269

RESUMEN

OBJECTIVE: The number of hematopoietic stem and progenitor cells (HPCs) per cord blood unit is limited, and this can result in delayed engraftment or graft failure. In vitro expansion of HPCs provides a perspective to overcome these limitations. Cytokines as well as mesenchymal stromal cells (MSCs) have been shown to support HPCs ex vivo expansion, but a systematic analysis of their interplay remains elusive. MATERIALS AND METHODS: Twenty different combinations of growth factors (stem cell factor [SCF], thrombopoietin [TPO], fibroblast growth factor-1 [FGF-1], angiopoietin-like 5, and insulin-like growth factor-binding protein 2), either with or without MSC coculture were systematically compared for their ability to support HPC expansion. CD34(+) cells were stained with carboxyfluorescein diacetate N-succinimidyl ester to monitor cell division history in conjunction with immunophenotype. Colony-forming unit frequencies and hematopoietic reconstitution of nonobese diabetic severe combined immunodeficient mice were also assessed. RESULTS: Proliferation of HPCs was stimulated by coculture with MSCs. This was further enhanced in combination with SCF, TPO, and FGF-1. Moreover, these conditions maintained expression of primitive surface markers for more than four cell divisions. Colony-forming unit-initiating cells were not expanded without stromal support, whereas an eightfold increase was reached by simultaneous cytokine-treatment and MSC coculture. Importantly, in comparison to expansion without stromal support, coculture with MSCs significantly enhanced hematopoietic chimerism in a murine transplantation model. CONCLUSIONS: The supportive effect of MSCs on hematopoiesis can be significantly increased by addition of specific recombinant growth factors; especially in combination with SCF, TPO, and FGF-1.


Asunto(s)
Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Mesodermo/citología , Células del Estroma/citología , Animales , Técnicas de Cocultivo , Sinergismo Farmacológico , Citometría de Flujo , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células del Estroma/inmunología
10.
Cytotherapy ; 12(7): 899-908, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20210674

RESUMEN

BACKGROUND: DNA and chromatin modifications are critical mediators in the establishment and maintenance of cell type-specific gene expression patterns that constitute cellular identities. One type of modification, the acetylation and deacetylation of histones, occurs reversibly on lysine ε-NH3(+) groups of core histones via histone acetyl transferases (HAT) and histone deacetylases (HDAC). Hyperacetylated histones are associated with active chromatin domains, whereas hypoacetylated histones are enriched in non-transcribed loci. METHODS: We analyzed global histone H4 acetylation and HDAC activity levels in mature lineage marker-positive (Lin(+)) and progenitor lineage marker-negative (Lin⁻) hematopoietic cells from murine bone marrow (BM). In addition, we studied the effects of HDAC inhibition on hematopoietic progenitor/stem cell (HPSC) frequencies, cell survival, differentiation and HoxB4 dependence. RESULTS: We observed that Lin⁻ and Lin(+) cells do not differ in global histone H4 acetylation but in HDAC activity levels. Further, we saw that augmented histone acetylation achieved by transient Trichostatin A (TSA) treatment increased the frequency of cells with HPSC immunophenotype and function in the heterogeneous pool of BM cells. Induction of histone hyperacetylation in differentiated BM cells was detrimental, as evidenced by preferential death of mature BM cells upon HDAC inhibition. Finally, TSA treatment of BM cells from HoxB4(-/-) mice revealed that the HDAC inhibitor-mediated increase in HPSC frequencies was independent of HoxB4. CONCLUSIONS: Overall, these data indicate the potential of chromatin modifications for the regulation of HPSC. Chromatin-modifying agents may provide potential strategies for ex vivo expansion of HPSC.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Acetilación/efectos de los fármacos , Animales , Antígenos de Diferenciación/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Linaje de la Célula , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Histona Desacetilasas/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética
11.
Mech Ageing Dev ; 131(2): 124-32, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20074583

RESUMEN

The potential use of neural stem cells in basic research, drug testing and for development of therapeutic strategies requires large scale in vitro amplification, increasing the probability of genetic instability and transformation. Little is known, however, about potential correlations between long-term culture of neural stem and progenitor cells (NSPCs), changed differentiation and self-renewal capacities, and the occurrence of chromosomal instability. This study investigates the effect of extended culture time on self-renewal, differentiation capacity, cell cycle phase distribution, telomere length, telomerase activity and chromosomal stability on fetal brain-derived cells that form floating sphere colonies (neurospheres). We observed that increased sphere-forming capacity indicative of increased proliferation was accompanied by a decreased ability to differentiate into neural lineages. The high mobility group A (Hmga2) gene positively regulates self-renewal via repression of p16(Ink4a) and p19(ARF) gene expression. This study discerned an upregulation of Hmga2 gene and protein expression and decreased p16(Ink4a) and p19(ARF) gene expression, suggesting that Hmga2 might promote the proliferation of neurosphere cells in long-term culture. Further, our analyses revealed a significant decrease in telomere length after 4 weeks of culturing that is paralleled by a moderate upregulation of telomerase activity. Importantly, regular gain of chromosome 1 with random structural chromosomal aberrations was observed within 16 weeks of neurosphere cell culture. Genetic instability and diminished differentiation capacity seem to be a consequence of long-term culture of neurosphere cells. These data indicate the necessity to analyze self-renewal, differentiation capacity, telomere length, tumor suppressor genes and chromosomal stability in neurosphere cultures prior to their usage in basic research, drug testing or the development of therapeutic strategies.


Asunto(s)
Ciclo Celular/genética , Diferenciación Celular/genética , Células Madre Multipotentes/metabolismo , Sistema Nervioso/metabolismo , Neuronas/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Embrión de Mamíferos/metabolismo , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso/embriología , Esferoides Celulares/metabolismo , Factores de Tiempo
12.
Int J Dev Biol ; 54(11-12): 1755-62, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21404193

RESUMEN

Patient derived stem cell-based therapies are considered a future treatment option for Parkinson´s disease, a chronic and progressive brain neurodegenerative disorder characterized by depletion of dopaminergic neurons in the basal ganglia. While many aspects of the in vitro and in vivo differentiation potential of uniparental parthenogenetic (PG) and gynogenetic (GG) embryonic stem (ES) cells of several species have been studied, the capacity of androgenetic (AG) ES cells to develop into neuronal subtypes remains unclear. Here, we investigated the potential of murine AG ES cells to undergo dopaminergic differentiation both via directed in vitro differentiation, and in vivo, in ES cell-chimeric E12.5 and E16.5 brains. We show that similar to normal (N; developed from a zygote with maternal and paternal genomes) ES cells, AG cells generated dopaminergic neurons in vitro and in E12.5 and E16.5 chimeric brains following blastocyst injection. Expression of brain-specific imprinted genes was maintained in AG and normal dopaminergic cell cultures. Our results indicate that AG ES cells have dopaminergic differentiation potential in vitro and in vivo. This contrasts with previous reports of limited neural in vivo differentiation of AG cells in later brain development, and suggests that AG ES cells could be therapeutically relevant for future cellular replacement strategies for brain disease.


Asunto(s)
Encéfalo/embriología , Diferenciación Celular , Dopamina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genoma , Neuronas/citología , Animales , Blastocisto , Encéfalo/citología , Línea Celular , Quimera/embriología , Quimera/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre , Cigoto
13.
Cells Tissues Organs ; 188(1-2): 103-15, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18600024

RESUMEN

Hematopoietic stem cells maintain the development of all mature blood cells throughout life due to their sustained self-renewal capacity and multilineage differentiation potential. During development into specific cell lineages, the options of stem cells and multipotent progenitor cells become increasingly restricted concomitant with a successive decline in self-renewal potential. Here we describe an Flt3+CD11b+ multipotent progenitor that can be amplified in vitro with a specific combination of cytokines to yield homogeneous populations in high cell numbers. By employing gene expression profiling with DNA microarrays, we studied the transcription factor repertoire of Flt3+CD11b+ progenitors and related it to the transcription factor repertoire of hematopoietic stem cells and embryonic stem cells. We report here on overlapping and nonoverlapping expression patterns of transcription factors in these cells and thus provide novel insights into the dynamic networks of transcriptional regulators in embryonic and adult stem cells. Additionally, the results obtained open the perspective for elucidating lineage and 'stemness' determinants in hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Línea Celular , Análisis por Conglomerados , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Factores de Transcripción/metabolismo
14.
Eur J Heart Fail ; 10(2): 119-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18279767

RESUMEN

OBJECTIVE: Bone marrow (BM)-derived haematopoietic stem cells have been proposed as a potential cell source to functionally engraft the myocardium and to improve cardiac function after myocardial infarction (MI). However, experimental and clinical data are inconsistent. Since the specific characteristics of different BM cell subsets could influence their therapeutic potential we determined the effect of different BM cell populations on left ventricular remodelling after MI. METHODS AND RESULTS: MI was induced in female mice by coronary artery ligation. Surviving mice were randomised to receive either: total BM, mature Lin(+) or primitive Lin(-) cells from male mice, or saline, via intracardiac injection. Injected cells were detected in the infarct and border zone by PCR for Y-chromosomal sequences. Serial transthoracic echocardiography was performed 1, 21, and 42 days after MI. Over a period of 6 weeks, mortality was not different between the groups. After MI, animals exhibited left ventricular dilatation, as expected. Left ventricular remodelling was not influenced by Lin(+) or Lin(-) BM cells but was partially improved by unfractionated BM cell injection. Paracrine secretion of cytokines (e.g. IL-6, GM-CSF) was differentially regulated in supernatants of cultured BM cells. SUMMARY: Treatment with unfractionated BM cells, but not Lin(+), or Lin(-) cells partially improved cardiac remodelling and function after MI. This may be mediated by paracrine effects.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , Remodelación Ventricular , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/mortalidad , Infarto del Miocardio/fisiopatología , Comunicación Paracrina/fisiología , Análisis de Supervivencia
15.
Stem Cells ; 26(4): 920-6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18203677

RESUMEN

Chromatin architecture in stem cells determines the pattern of gene expression and thereby cell identity and fate. The chromatin-modifying agents trichostatin A (TSA) and 5-Aza-2'-deoxycytidine (AzaC) affect histone acetylation and DNA methylation, respectively, and thereby influence chromatin structure and gene expression. In our previous work, we demonstrated that TSA/AzaC treatment of neurosphere cells induces hematopoietic activity in vivo that is long-term, multilineage, and transplantable. Here, we have analyzed the TSA/AzaC-induced changes in gene expression by global gene expression profiling. TSA/AzaC caused both up- and downregulation of genes, without increasing the total number of expressed genes. Chromosome analysis showed no hot spot of TSA/AzaC impact on a particular chromosome or chromosomal region. Hierarchical cluster analysis revealed common gene expression patterns among neurosphere cells treated with TSA/AzaC, embryonic stem (ES) cells, and hematopoietic stem cells. Furthermore, our analysis identified several stem cell genes and pluripotency-associated genes that are induced by TSA/AzaC in neurosphere cells, including Cd34, Cd133, Oct4, Nanog, Klf4, Bex1, and the Dppa family members Dppa2, 3, 4, and 5. Sox2 and c-Myc are constitutively expressed in neurosphere cells. We propose a model in which TSA/AzaC, by removal of epigenetic inhibition, induces the reactivation of several stem cell and pluripotency-associated genes, and their coordinate expression enlarges the differentiation potential of somatic precursor cells.


Asunto(s)
Cromatina/genética , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/fisiología , Células Madre Pluripotentes/fisiología , Animales , Células Cultivadas , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/fisiología
16.
Organogenesis ; 4(1): 33-41, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19279713

RESUMEN

The biological role of genomic imprinting in adult tissue is central to the consideration of transplanting uniparental embryonic stem (ES) cell-derived tissues. We have recently shown that both maternal (parthenogenetic/gynogenetic) and paternal (androgenetic) uniparental ES cells can differentiate, both in vivo in chimeras and in vitro, into adult-repopulating hematopoietic stem and progenitor cells. This suggests that, at least in some tissues, the presence of two maternal or two paternal genomes does not interfere with stem cell function and tissue homeostasis in the adult. Here, we consider implications of the contribution of uniparental cells to hematopoiesis and to development of other organ systems, notably neural tissue for which consequences of genomic imprinting are associated with a known bias in development and behavioral disorders. Our findings so far indicate that there is little or no limit to the differentiation potential of uniparental ES cells outside the normal developmental paradigm. As a potentially donor MHC-matching source of tissue, uniparental transplants may provide not only a clinical resource but also a unique tool to investigate aspects of genomic imprinting in adults.

17.
Leuk Res ; 32(5): 781-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17980910

RESUMEN

While critical steps in the regulation of leukemia cell development have been intensively studied in recent years, less is known about the interactions of leukemic cells with their stroma. Previously, we have shown that human acute myeloid leukemia (AML) cells differentiate upon injection into murine blastocysts. We here describe that human AML Kasumi-1 cells, cocultured with murine aorta-gonad-mesonephros (AGM) region-derived DAS104-4 stromal cells, decrease proliferation and colony formation efficiency; and up-regulate myelo-monocytic cell surface markers. Gene expression analysis showed decreased transcription of the AML1-ETO fusion gene and increased transcription of p16 (INK4A), p21 (WAF1) and C/EBPalpha genes. Coculture can induce myeloid differentiation also in patient-derived AML cells. Our findings strengthen the notion that the embryonic milieu can regulate the proliferation and differentiation of leukemic cells.


Asunto(s)
Aorta/embriología , Gónadas/embriología , Leucemia Mieloide Aguda/patología , Mesonefro/embriología , Células del Estroma/fisiología , Aorta/citología , Antígeno CD11b/análisis , Diferenciación Celular , Línea Celular , Proliferación Celular , Técnicas de Cocultivo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Gónadas/citología , Humanos , Receptores de Lipopolisacáridos/análisis , Mesonefro/citología , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1
18.
Mech Ageing Dev ; 127(7): 600-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16620920

RESUMEN

BCL-2 functions as a death repressor molecule in an evolutionary conserved cell death pathway. Inactivation of bcl-2 in mice results in pleiotropic effects including postnatal growth retardation, massive apoptosis in lymphoid tissues, polycystic kidney disease (PKD) and shortened lifespan. To evaluate the influence of the affected bcl-2 deficient kidneys on the postnatal development and lifespan of bcl-2 knockout mice we used "the rescue of (n-1) affected tissues" strategy. According to this strategy bcl-2 heterozygous animals were crossed with H2K-hbcl-2 transgenic mice expressing human BCL-2 in most tissues and organs excluding the kidney. Overexpression of hBCL-2 in bcl-2-/- mice rescues growth retardation, normalizes and protects the hematolymphoid system from gamma-radiation. However, the hbcl-2 transgene is not expressed in kidneys and the rescued mice have PKD and a shortened lifespan. Thus, our results indicated that PKD is the main reason of early mortality in bcl-2 deficient mice. Moreover, we have created mouse model, similar to the kidney specific knockout of bcl-2. Such models can be useful to study the influence of bcl-2 or other gene deficiency in individual organs (or tissues) on development and ageing of whole organism.


Asunto(s)
Apoptosis/genética , Longevidad/genética , Enfermedades Renales Poliquísticas/genética , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Insuficiencia Renal/genética , Animales , Cruzamientos Genéticos , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/patología , Heterocigoto , Humanos , Ratones , Ratones Noqueados , Enfermedades Renales Poliquísticas/patología , Insuficiencia Renal/patología
19.
Leuk Res ; 29(10): 1191-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15941586

RESUMEN

To better characterize acute myeloid leukemia (AML) development in non-obese diabetic (NOD)/severe combined immunodeficiency (SCID) mice, we transplanted samples from patients with AML or KG-1 and EOL-1 cell lines. We found 9/12 primary AML samples and both cell lines to engraft within 2-8 weeks, with 5-80% human cells in bone marrow. Compared with freshly isolated AML cells, percentages of human CD33+, CD38+, CD31+ CD13+ or CD15+ subpopulations increased after transplantation, whereas percentages of CD34+ cells decreased. Engrafted mice frequently showed expression of human endothelial cell markers. Thus, transplantation of human AML into NOD/SCID mice reveals expression of hematopoietic and endothelial differentiation markers.


Asunto(s)
Antígenos de Superficie/metabolismo , Biomarcadores de Tumor/metabolismo , Leucemia Mieloide/patología , Trasplante Heterólogo/patología , Enfermedad Aguda , Adulto , Anciano , Animales , Endotelio Vascular/citología , Endotelio Vascular/embriología , Endotelio Vascular/patología , Citometría de Flujo , Humanos , Leucemia Mieloide/clasificación , Leucemia Mieloide/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo/inmunología , Células Tumorales Cultivadas
20.
J Immunol ; 174(5): 2552-62, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728461

RESUMEN

Dendritic cells (DC) represent key regulators of the immune system, yet their development from hemopoietic precursors is poorly defined. In this study, we describe an in vitro system for amplification of a Flt3(+)CD11b(+) progenitor from mouse bone marrow with specific cytokines. Such progenitor cells develop into both CD11b(+) and CD11b(-) DC, and CD8alpha(+) and CD8alpha(-) DC in vivo. Furthermore, with GM-CSF, these progenitors synchronously differentiated into fully functional DC in vitro. This two-step culture system yields homogeneous populations of Flt3(+)CD11b(+) progenitor cells in high numbers and allows monitoring the consecutive steps of DC development in vitro under well-defined conditions. We used phenotypic and functional markers and transcriptional profiling by DNA microarrays to study the Flt3(+)CD11b(+) progenitor and differentiated DC. We report here on an extensive analysis of the surface Ag expression of Flt3(+)CD11b(+) progenitor cells and relate that to surface Ag expression of hemopoietic stem cells. Flt3(+)CD11b(+) progenitors studied exhibit a broad overlap of surface Ags with stem cells and express several stem cell Ags such as Flt3, IL-6R, c-kit/SCF receptor, and CD93/AA4.1, CD133/AC133, and CD49f/integrin alpha(6). Thus, Flt3(+)CD11b(+) progenitors express several stem cell surface Ags and develop into both CD11b(+) and CD11b(-) DC, and CD8alpha(+) and CD8alpha(-) DC in vivo, and thus into both of the main conventional DC subtypes.


Asunto(s)
Antígeno CD11b/biosíntesis , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Madre Hematopoyéticas/citología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Traslado Adoptivo , Secuencia de Aminoácidos , Animales , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Tirosina Quinasa 3 Similar a fms
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA