Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 136(21): 2410-2415, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32599615

RESUMEN

Although cytokine-mediated expansion of human hematopoietic stem cells (HSCs) can result in high yields of hematopoietic progenitor cells, this generally occurs at the expense of reduced bone marrow HSC repopulating ability, thereby limiting potential therapeutic applications. Because bromodomain-containing proteins (BCPs) have been demonstrated to regulate mouse HSC self-renewal and stemness, we screened small molecules targeting various BCPs as potential agents for ex vivo expansion of human HSCs. Of 10 compounds tested, only the bromodomain and extra-terminal motif inhibitor CPI203 enhanced the expansion of human cord blood HSCs without losing cell viability in vitro. The expanded cells also demonstrated improved engraftment and repopulation in serial transplantation assays. Transcriptomic and functional studies showed that the expansion of long-term repopulating HSCs was accompanied by synchronized expansion and maturation of megakaryocytes consistent with CPI203-mediated reprogramming of cord blood hematopoietic stem and progenitor cells. This approach may therefore prove beneficial for ex vivo gene editing, for enhanced platelet production, and for the improved usage of cord blood for transplantation research and therapy.


Asunto(s)
Acetamidas/farmacología , Azepinas/farmacología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Proteínas/antagonistas & inhibidores , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Supervivencia de Injerto/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos NOD , Transcriptoma/efectos de los fármacos
2.
Int J Mol Sci ; 23(7)2022 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-35409034

RESUMEN

The past five decades have seen significant progress in our understanding of human hematopoiesis. This has in part been due to the unprecedented development of advanced technologies, which have allowed the identification and characterization of rare subsets of human hematopoietic stem and progenitor cells and their lineage trajectories from embryonic through to adult life. Additionally, surrogate in vitro and in vivo models, although not fully recapitulating human hematopoiesis, have spurred on these scientific advances. These approaches have heightened our knowledge of hematological disorders and diseases and have led to their improved diagnosis and therapies. Here, we review human hematopoiesis at each end of the age spectrum, during embryonic and fetal development and on aging, providing exemplars of recent progress in deciphering the increasingly complex cellular and molecular hematopoietic landscapes in health and disease. This review concludes by highlighting links between chronic inflammation and metabolic and epigenetic changes associated with aging and in the development of clonal hematopoiesis.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Envejecimiento/genética , Hematopoyesis Clonal , Epigénesis Genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos
3.
Blood ; 134(13): 1059-1071, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31383639

RESUMEN

Human lymphopoiesis is a dynamic lifelong process that starts in utero 6 weeks postconception. Although fetal B-lymphopoiesis remains poorly defined, it is key to understanding leukemia initiation in early life. Here, we provide a comprehensive analysis of the human fetal B-cell developmental hierarchy. We report the presence in fetal tissues of 2 distinct CD19+ B-progenitors, an adult-type CD10+ve ProB-progenitor and a new CD10-ve PreProB-progenitor, and describe their molecular and functional characteristics. PreProB-progenitors and ProB-progenitors appear early in the first trimester in embryonic liver, followed by a sustained second wave of B-progenitor development in fetal bone marrow (BM), where together they form >40% of the total hematopoietic stem cell/progenitor pool. Almost one-third of fetal B-progenitors are CD10-ve PreProB-progenitors, whereas, by contrast, PreProB-progenitors are almost undetectable (0.53% ± 0.24%) in adult BM. Single-cell transcriptomics and functional assays place fetal PreProB-progenitors upstream of ProB-progenitors, identifying them as the first B-lymphoid-restricted progenitor in human fetal life. Although fetal BM PreProB-progenitors and ProB-progenitors both give rise solely to B-lineage cells, they are transcriptionally distinct. As with their fetal counterparts, adult BM PreProB-progenitors give rise only to B-lineage cells in vitro and express the expected B-lineage gene expression program. However, fetal PreProB-progenitors display a distinct, ontogeny-related gene expression pattern that is not seen in adult PreProB-progenitors, and they share transcriptomic signatures with CD10-ve B-progenitor infant acute lymphoblastic leukemia blast cells. These data identify PreProB-progenitors as the earliest B-lymphoid-restricted progenitor in human fetal life and suggest that this fetal-restricted committed B-progenitor might provide a permissive cellular context for prenatal B-progenitor leukemia initiation.


Asunto(s)
Feto/citología , Linfopoyesis , Neprilisina/análisis , Células Precursoras de Linfocitos B/citología , Adulto , Médula Ósea/embriología , Médula Ósea/metabolismo , Células Cultivadas , Feto/embriología , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Hígado/embriología , Hígado/metabolismo , Neprilisina/genética , Células Precursoras de Linfocitos B/metabolismo , Transcriptoma
4.
Transfusion ; 59(12): 3560-3569, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31769050

RESUMEN

BACKGROUND: Umbilical cord blood (UCB) is a source of hematopoietic stem cells for transplantation, offering an alternative for patients unable to find a matched adult donor. UCB is also a versatile source of hematopoietic stem and progenitor cells (hCD34 + HSPCs) for research into hematologic diseases, in vitro expansion, ex vivo gene therapy, and adoptive immunotherapy. For these studies, there is a need to isolate hCD34 + HSPCs from cryopreserved units, and protocols developed for isolation from fresh cord blood are unsuitable. STUDY DESIGN: This study describes a modified method for isolating hCD34 + HSPCs from cryopreserved UCB. It uses the Plasmatherm system for thawing, followed by CD34 microbead magnetic-activated cell sorting isolation with a cell separation kit (Whole Blood Columns, Miltenyi Biotec). hCD34 + HSPC phenotypes and functionality were assessed in vitro and hematologic reconstitution determined in vivo in immunodeficient mice. RESULTS: Total nucleated cell recovery after thawing and washing was 44.7 ± 11.7%. Recovery of hCD34 + HSPCs after application of thawed cells to Whole Blood Columns was 77.5 ± 22.6%. When assessed in two independent laboratories, the hCD34+ cell purities were 71.7 ± 10.7% and 87.8 ± 2.4%. Transplantation of the enriched hCD34 + HSPCs into NSG mice revealed the presence of repopulating hematopoietic stem cells (estimated frequency of 0.07%) and multilineage engraftment. CONCLUSION: This provides a simplified protocol for isolating high-purity human CD34 + HSPCs from banked UCB adaptable to current Good Manufacturing Practice. This protocol reduces the number of steps and associated risks and thus total production costs. Importantly, the isolated CD34 + HSPCs possess in vivo repopulating activity in immunodeficient mice, making them a suitable starting population for ex vivo culture and gene editing.


Asunto(s)
Antígenos CD34/metabolismo , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Células Madre/citología , Animales , Criopreservación , Edición Génica , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoterapia , Ratones , Células Madre/metabolismo
5.
Stem Cells ; 34(6): 1664-78, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26866290

RESUMEN

Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1ß activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Molécula A de Adhesión de Unión/metabolismo , Receptores CXCR4/metabolismo , Antígeno AC133/metabolismo , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Células HL-60 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Jurkat , Unión Proteica/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos
7.
Transfusion ; 56(4): 899-904, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26662223

RESUMEN

BACKGROUND: Processing of bone marrow (BM) is often required to remove incompatible red blood cells (RBCs) or to reduce the volume before transplantation or cryopreservation. We have evaluated the Spectra Optia apheresis system to determine its effectiveness in volume reduction and RBC depletion of human BM before transplantation. STUDY DESIGN AND METHODS: BM from 30 donations (28 allogeneic and two autologous) were processed using the Spectra Optia over a 12-month period. The mean BM collection volume was 1094 ± 337 mL and RBC volume was 374 ± 148 mL. Processing using the Spectra Optia was as described by the manufacturer. RESULTS: Volume reduction achieved was 93.0 ± 1.2%; RBC depletion was 98.8 ± 0.4%; and mononuclear, CD34+, and CD3+ cell recoveries were 79.12 ± 14.03, 88.36 ± 13.76, and 79.84 ± 16.27%, respectively. In total 26 of 28 processed allografts were transplanted; 24 achieved neutrophil engraftment in 20.7 ± 5.9 days and 18 achieved platelet engraftment in 19.6 ± 8.9 days. Time in transit significantly affected the Spectra Optia's ability to recover mononuclear, CD34+, and CD3+ cells, and the overall age of the BM at the time of processing significantly affected the recovery of mononuclear and CD3+ cells, but not CD34+ cells. Time in storage at 2 to 6°C had no adverse effect on processing. CONCLUSION: This study demonstrates that the Spectra Optia can effectively volume reduce and RBC deplete human BM before transplantation. Time in transit should be as short as possible but may be extended up to 24 hours if the donation is refrigerated during transit.


Asunto(s)
Eliminación de Componentes Sanguíneos/instrumentación , Células de la Médula Ósea/citología , Antígenos CD34/metabolismo , Eliminación de Componentes Sanguíneos/métodos , Conservación de la Sangre/métodos , Conservación de la Sangre/normas , Recolección de Muestras de Sangre/métodos , Recolección de Muestras de Sangre/normas , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Separación Celular/instrumentación , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Trasplante de Células Madre Hematopoyéticas , Humanos , Temperatura , Factores de Tiempo , Trasplante Homólogo , Transportes
8.
Br J Haematol ; 169(4): 552-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25757087

RESUMEN

Murine models of bone marrow transplantation show that pre-conditioning regimens affect the integrity of the bone marrow endothelium and that the repair of this vascular niche is an essential pre-requisite for successful haematopoietic stem and progenitor cell engraftment. Little is known about the angiogenic pathways that play a role in the repair of the human bone marrow vascular niche. We therefore established an in vitro humanized model, composed of bone marrow stromal and endothelial cells and have identified several pro-angiogenic factors, VEGFA, ANGPT1, CXCL8 and CXCL16, produced by the stromal component of this niche. We demonstrate for the first time that addition of CXCL8 or inhibition of its receptor, CXCR2, modulates blood vessel formation in our bone marrow endothelial niche model. Compared to wild type, Cxcr2(-/-) mice displayed a reduction in bone marrow cellularity and delayed platelet and leucocyte recovery following myeloablation and bone marrow transplantation. The delay in bone marrow recovery correlated with impaired bone marrow vascular repair. Taken together, our data demonstrate that CXCR2 regulates bone marrow blood vessel repair/regeneration and haematopoietic recovery, and clinically may be a therapeutic target for improving bone marrow transplantation.


Asunto(s)
Trasplante de Médula Ósea , Médula Ósea/irrigación sanguínea , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Neovascularización Fisiológica , Receptores de Interleucina-8B/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Línea Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Receptores de Interleucina-8B/genética , Acondicionamiento Pretrasplante
9.
Cytotherapy ; 17(9): 1268-79, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26276008

RESUMEN

BACKGROUND AIMS: There is real and sustained interest in preparing T-regulatory cells from leukapheresis collections for cellular therapy through the use of simple, effective and reliable methods conforming to Good Manufacturing Practice (GMP). We describe a GMP-ready isolation procedure for CD25(+) products with the use of a sterile docking device, pigtail sampling, a laminar flow hood and the CliniMACS system that uses leukapheresis collections made by two apheresis machines. METHODS: We used CD8/CD19 depletion followed by CD25-positive selection. The median number of CD4(+) cells recovered was 72.5 ± 32.6 × 10(6), of which 60.5% ± 17.8% were CD25(+)FOXP3(+) cells. Suppression of autologous CD25(-) cell proliferation by the cryopreserved CD25(+) products was 51.3% ± 4.4%, 49.0% ± 3.7% and 39.0% ± 3.6% at CD25(+):CD25(-) ratios of 1:1, 1:2 and 1:4 (n = 6), respectively, comparable to suppression by fresh CD25(+) products (53% ± 6.2%, 51% ± 3.3% and 39% ± 7.1%). RESULTS: We found Leukapheresis collections by Cobe Spectra contained more lymphocytes and platelets than collections by Spectra Optia apheresis machine (median, 9.2 × 10(9) versus 6.7 × 10(9); P = 0.04) and platelets (median, 610 × 10(9) versus 170 × 10(9); P = 0.04). The frequency of CD4(+)CD25(+)FOXP3(+) was significantly higher in the leukapheresis (4.85%; 95% confidence interval, 1.95% to 5.38%) than in peripheral blood (3.9%; 95% confidence interval, 2.63% to 6.45%) (P = 0.02). The CD8- and CD19-negative depletion step was associated with significant loss of total CD4(+) T cells (P = 0.001). CONCLUSIONS: Results suggest that functional CD25(+) products can be isolated with a GMP-ready method, and good recovery can be obtained with the use of an optimized cryopreservation protocol. These data and methods show the potential, possibilities and future work needed to isolate target cell populations in a reproducible, time-efficient and cost-efficient manner for clinical applications.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Leucaféresis/métodos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/trasplante , Adulto , Proliferación Celular , Criopreservación/métodos , Humanos , Inmunofenotipificación , Recuento de Linfocitos , Depleción Linfocítica/métodos , Persona de Mediana Edad , Donantes de Tejidos
10.
Br J Haematol ; 166(3): 435-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837254

RESUMEN

Human induced pluripotent stem cells (hiPSCs), like embryonic stem cells, are under intense investigation for novel approaches to model disease and for regenerative therapies. Here, we describe the derivation and characterization of hiPSCs from a variety of sources and show that, irrespective of origin or method of reprogramming, hiPSCs can be differentiated on OP9 stroma towards a multi-lineage haemo-endothelial progenitor that can contribute to CD144(+) endothelium, CD235a(+) erythrocytes (myeloid lineage) and CD19(+) B lymphocytes (lymphoid lineage). Within the erythroblast lineage, we were able to demonstrate by single cell analysis (flow cytometry), that hiPSC-derived erythroblasts express alpha globin as previously described, and that a sub-population of these erythroblasts also express haemoglobin F (HbF), indicative of fetal definitive erythropoiesis. More notably however, we were able to demonstrate that a small sub-fraction of HbF positive erythroblasts co-expressed HbA in a highly heterogeneous manner, but analogous to cord blood-derived erythroblasts when cultured using similar methods. Moreover, the HbA expressing erythroblast population could be greatly enhanced (44·0 ± 6·04%) when a defined serum-free approach was employed to isolate a CD31(+) CD45(+) erythro-myeloid progenitor. These findings demonstrate that hiPSCs may represent a useful alternative to standard sources of erythrocytes (RBCs) for future applications in transfusion medicine.


Asunto(s)
Eritroblastos/citología , Eritroblastos/metabolismo , Eritropoyesis/fisiología , Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Globinas beta/genética , gamma-Globinas/genética , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Linaje de la Célula , Variaciones en el Número de Copia de ADN , Humanos , Inmunofenotipificación , Cariotipificación , Globinas beta/metabolismo , gamma-Globinas/metabolismo
11.
Cancer Immunol Immunother ; 63(2): 133-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24190544

RESUMEN

Adoptive cell therapy employing gene-modified T-cells expressing chimeric antigen receptors (CARs) has shown promising preclinical activity in a range of model systems and is now being tested in the clinical setting. The manufacture of CAR T-cells requires compliance with national and European regulations for the production of medicinal products. We established such a compliant process to produce T-cells armed with a first-generation CAR specific for carcinoembryonic antigen (CEA). CAR T-cells were successfully generated for 14 patients with advanced CEA(+) malignancy. Of note, in the majority of patients, the defined procedure generated predominantly CD4(+) CAR T-cells with the general T-cell population bearing an effector-memory phenotype and high in vitro effector function. Thus, improving the process to generate less-differentiated T-cells would be more desirable in the future for effective adoptive gene-modified T-cell therapy. However, these results confirm that CAR T-cells can be generated in a manner compliant with regulations governing medicinal products in the European Union.


Asunto(s)
Traslado Adoptivo , Antígeno Carcinoembrionario/inmunología , Proteínas Quimerinas/biosíntesis , Receptores de Antígenos de Linfocitos T/biosíntesis , Linfocitos T/inmunología , Humanos , Inmunofenotipificación , Interferón gamma/biosíntesis
12.
Transfusion ; 54(5): 1278-85, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24224530

RESUMEN

BACKGROUND: Cryopreserved umbilical cord blood (CB) is increasingly used as a cell source to reconstitute marrow in hematopoietic stem cell transplant patients. Delays in cryopreservation may adversely affect cell viability, thereby reducing their potential for engraftment after transplantation. STUDY DESIGN AND METHODS: The impact of delayed cryopreservation for up to 3 days on the viability of both CD45+ and CD34+ cell populations in 28 CB donations with volumes of 58.40 ± 15.4 mL (range, 39.4-107.4 mL) was investigated to establish whether precryopreservation storage time could be extended from our current time of 24 to 48 hours in line with other CB banks. Viability was assessed on 3 consecutive days, both before and after cryopreservation, by flow cytometry using 7-aminoactinomycin D (7-AAD) and annexin V methods. RESULTS: The results using 7-AAD and annexin V indicated the viability of CD34+ cells before cryopreservation remained high (>92.33 ± 4.11%) over 3 days, whereas the viability of CD45+ cells decreased from 86.36 ± 4.97% to 66.24 ± 7.78% (p < 0.0001) by Day 3. Storage time significantly affected the viability of CD34+ cells after cryopreservation. Using 7-AAD, the mean CD34+ cell viability decreased by approximately 5% per extra day in storage from 84.30 ± 6.27% on Day 1 to 79.01 ± 7.44% (p < 0.0057) on Day 2 and to 73.95 ± 7.54% (p < 0.0001) on Day 3. With annexin V staining CD34+ cell viability fell by approximately 7% per extra day in storage from 77.17 ± 8.47% on Day 1 to 69.56 ± 13.30% (p < 0.0194) on Day 2 and to 62.89 ± 15.22% (p < 0.0002) on Day 3. CONCLUSION: This study demonstrates that extended precryopreservation storage adversely affects viability and should be avoided.


Asunto(s)
Conservación de la Sangre , Sangre Fetal/citología , Anexina A5/análisis , Antígenos CD34/análisis , Supervivencia Celular , Criopreservación , Dactinomicina/análogos & derivados , Dactinomicina/análisis , Humanos , Antígenos Comunes de Leucocito/análisis , Factores de Tiempo
13.
Br Med Bull ; 108: 25-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24152971

RESUMEN

BACKGROUND: Blood vessel formation is fundamental to development, while its dysregulation can contribute to serious disease. Expectations are that hundreds of millions of individuals will benefit from therapeutic developments in vascular biology. MSCs are central to the three main vascular repair mechanisms. SOURCES OF DATA: Key recent published literature and ClinicalTrials.gov. AREAS OF AGREEMENT: MSCs are heterogeneous, containing multi-lineage stem and partly differentiated progenitor cells, and are easily expandable ex vivo. There is no single marker defining native MSCs in vivo. Their phenotype is strongly determined by their specific microenvironment. Bone marrow MSCs have skeletal stem cell properties. Having a perivascular/vascular location, they contribute to vascular formation and function and might be harnessed to regenerate a blood supply to injured tissues. AREAS OF CONTROVERSY: These include MSC origin, phenotype and location in vivo and their ability to differentiate into functional cardiomyocytes and endothelial cells or act as vascular stem cells. In addition their efficacy, safety and potency in clinical trials in relation to cell source, dose, delivery route, passage and timing of administration, but probably even more on the local preconditioning and the mechanisms by which they exert their effects. GROWING POINTS: Understanding the origin and the regenerative environment of MSCs, and manipulating their homing properties, proliferative ability and functionality through drug discovery and reprogramming strategies are important for their efficacy in vascular repair for regenerative medicine therapies and tissue engineering approaches. AREAS TIMELY FOR DEVELOPING RESEARCH: Characterization of MSCs' in vivo origins and biological properties in relation to their localization within tissue niches, reprogramming strategies and newer imaging/bioengineering approaches.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Medicina Regenerativa/métodos , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Enfermedades Cardiovasculares/terapia , Quimiocinas/metabolismo , Humanos , Microvasos/crecimiento & desarrollo , Enfermedades de la Piel/terapia , Ingeniería de Tejidos/métodos
14.
Blood ; 117(15): 4008-11, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21343609

RESUMEN

Induced pluripotent stem (iPS) cells offer a unique potential for understanding the molecular basis of disease and development. Here we have generated several human iPS cell lines, and we describe their pluripotent phenotype and ability to differentiate into erythroid cells, monocytes, and endothelial cells. More significantly, however, when these iPS cells were differentiated under conditions that promote lympho-hematopoiesis from human embryonic stem cells, we observed the formation of pre-B cells. These cells were CD45(+)CD19(+)CD10(+) and were positive for transcripts Pax5, IL7αR, λ-like, and VpreB receptor. Although they were negative for surface IgM and CD5 expression, iPS-derived CD45(+)CD19(+) cells also exhibited multiple genomic D-J(H) rearrangements, which supports a pre-B-cell identity. We therefore have been able to demonstrate, for the first time, that human iPS cells are able to undergo hematopoiesis that contributes to the B-cell lymphoid lineage.


Asunto(s)
Linfocitos B/citología , Linfopoyesis/fisiología , Células Madre Pluripotentes/citología , Células Precursoras de Linfocitos B/citología , Adulto , Antígenos CD19/metabolismo , Linfocitos B/fisiología , Línea Celular , Linaje de la Célula/inmunología , Humanos , Inmunoglobulina de Cadenas Ligeras Subrogadas/genética , Inmunofenotipificación , Antígenos Comunes de Leucocito/metabolismo , Neprilisina/metabolismo , Factor de Transcripción PAX5/genética , Células Madre Pluripotentes/fisiología , Células Precursoras de Linfocitos B/fisiología , Receptores de Interleucina-7/genética
15.
Br J Haematol ; 157(3): 299-311, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22324374

RESUMEN

The bone marrow contains specific microenvironmental stem cell niches that maintain haemopoiesis. CXCL12-expressing mesenchymal stromal cells are closely associated with the bone marrow sinusoidal endothelia, forming key elements of the haemopoietic stem cell niche, yet their ability to regulate endothelial function is not clearly defined. Given that the murine nestin(+) cell line, MS-5, provides a clonal surrogate bone marrow stromal niche capable of regulating both murine and human primitive haemopoietic stem/progenitor cell (HSC/HPC) fate in vitro, we hypothesized that MS-5 cells might also support new blood vessel formation and function. Here, for the first time, we demonstrate that this is indeed the case. Using proteome arrays, we identified HSC/HPC active angiogenic factors that are preferentially secreted by haemopoietic supportive nestin(+) MS-5 cells, including CXCL12 (SDF-1), NOV (CCN3), HGF, Angiopoietin-1 and CCL2 (MCP-1). Concentrating on CXCL12, we confirmed its presence in MS-5 conditioned media and demonstrated that its antagonist in receptor binding, AMD-3100, which mobilizes HSC/HPCs and endothelial progenitors from bone marrow, could significantly reduce MS-5 mediated human vasculogenesis in vitro, principally by regulating human endothelial cell migration. Thus, the clonal nestin(+) MS-5 murine bone marrow stromal cell line not only promotes human haemopoiesis but also induces human vasculogenesis, with CXCL12 playing important roles in both processes.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Inductores de la Angiogénesis/metabolismo , Animales , Células de la Médula Ósea/fisiología , Comunicación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Proliferación Celular , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/fisiología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos , Ratones , Proteómica/métodos
16.
Biomater Transl ; 3(1): 31-54, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35837343

RESUMEN

Haematopoietic microenvironmental niches have been described as the 'gatekeepers' for the blood and immune systems. These niches change during ontogeny, with the bone marrow becoming the predominant site of haematopoiesis in post-natal life under steady state conditions. To determine the structure and function of different haematopoietic microenvironmental niches, it is essential to clearly define specific haematopoietic stem and progenitor cell subsets during ontogeny and to understand their temporal appearance and anatomical positioning. A variety of haematopoietic and non-haematopoietic cells contribute to haematopoietic stem and progenitor cell niches. The latter is reported to include endothelial cells and mesenchymal stromal cells (MSCs), skeletal stem cells and/or C-X-C motif chemokine ligand 12-abundant-reticular cell populations, which form crucial components of these microenvironments under homeostatic conditions. Dysregulation or deterioration of such cells contributes to significant clinical disorders and diseases worldwide and is associated with the ageing process. A critical appraisal of these issues and of the roles of MSC/C-X-C motif chemokine ligand 12-abundant-reticular cells and the more recently identified skeletal stem cell subsets in bone marrow haematopoietic niche function under homeostatic conditions and during ageing will form the basis of this research review. In the context of haematopoiesis, clinical translation will deal with lessons learned from the vast experience garnered from the development and use of MSC therapies to treat graft versus host disease in the context of allogeneic haematopoietic transplants, the recent application of these MSC therapies to treating emerging and severe coronavirus disease 2019 (COVID-19) infections, and, given that skeletal stem cell ageing is one proposed driver for haematopoietic ageing, the potential contributions of these stem cells to haematopoiesis in healthy bone marrow and the benefits and challenges of using this knowledge for rejuvenating the age-compromised bone marrow haematopoietic niches and restoring haematopoiesis.

17.
Angiogenesis ; 14(3): 381-92, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21720855

RESUMEN

As umbilical cord blood (UCB) is a rich source of endothelial colony-forming cells (ECFC), our aim was twofold: (1) to examine potential obstetric selection criteria for achieving the highest ECFC yields from UCB units, and (2) to determine whether transient storage temperatures of fresh UCB and cryopreservation of UCB units affected ECFC yield and function. ECFC quality was assessed before and after cryopreservation by their clonogenic proliferative potential. Of the 20 factors examined, placental weight was the only statistically significant obstetric factor that predicted ECFC frequency in UCB. Studies on the effects of storage revealed that transient storage of fresh UCB at 4°C reduced ECFC yield compared with storage at 22°C, while cryopreservation of UCB MNCs significantly reduced ECFC recoveries. To our knowledge, this is the first demonstration that placental weight and temperature of storage prior to processing or culture have significant effects on ECFC frequency in UCB. Our studies further support the evidence that cryopreservation of UCB MNCs compromises ECFC recovery.


Asunto(s)
Conservación de la Sangre , Criopreservación , Parto Obstétrico , Células Endoteliales/citología , Sangre Fetal/citología , Células Madre/citología , Células Endoteliales/metabolismo , Femenino , Sangre Fetal/metabolismo , Humanos , Recién Nacido , Masculino , Tamaño de los Órganos , Placenta/citología , Placenta/metabolismo , Embarazo , Células Madre/metabolismo , Factores de Tiempo
18.
NPJ Regen Med ; 6(1): 33, 2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103536

RESUMEN

Accurately defining hierarchical relationships between human stem cells and their progeny, and using this knowledge for new cellular therapies, will undoubtedly lead to further successful treatments for life threatening and chronic diseases, which represent substantial burdens on patient quality of life and to healthcare systems globally. Clinical translation relies in part on appropriate biomarker, in vitro manipulation and transplantation strategies. CD164 has recently been cited as an important biomarker for enriching both human haematopoietic and skeletal stem cells, yet a thorough description of extant human CD164 monoclonal antibody (Mab) characteristics, which are critical for identifying and purifying these stem cells, was not discussed in these articles. Here, we highlight earlier but crucial research describing these relevant characteristics, including the differing human CD164 Mab avidities and their binding sites on the human CD164 sialomucin, which importantly may affect subsequent stem cell function and fate.

19.
Front Immunol ; 12: 642198, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868276

RESUMEN

Humanized immune system (HIS) mouse models are useful tools for the in vivo investigation of human hematopoiesis. However, the majority of HIS models currently in use are biased towards lymphocyte development and fail to support long-term multilineage leucocytes and erythrocytes. Those that achieve successful multilineage reconstitution often require preconditioning steps which are expensive, cause animal morbidity, are technically demanding, and poorly reproducible. In this study, we address this challenge by using HSPC-NBSGW mice, in which NOD,B6.SCID IL-2rγ-/-KitW41/W41 (NBSGW) mice are engrafted with human CD133+ hematopoietic stem and progenitor cells (HSPCs) without the need for preconditioning by sublethal irradiation. These HSPCs are enriched in long-term hematopoietic stem cells (LT-HSCs), while NBSGW mice are permissive to human hematopoietic stem cell (HSC) engraftment, thus reducing the cell number required for successful HIS development. B cells reconstitute with the greatest efficiency, including mature B cells capable of class-switching following allogeneic stimulation and, within lymphoid organs and peripheral blood, T cells at a spectrum of stages of maturation. In the thymus, human thymocytes are identified at all major stages of development. Phenotypically distinct subsets of myeloid cells, including dendritic cells and mature monocytes, engraft to a variable degree in the bone marrow and spleen, and circulate in peripheral blood. Finally, we observe human erythrocytes which persist in the periphery at high levels following macrophage clearance. The HSPC-NBSGW model therefore provides a useful platform for the study of human hematological and immunological processes and pathologies.


Asunto(s)
Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Xenoinjertos , Modelos Animales , Animales , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID
20.
Cells ; 9(5)2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32365526

RESUMEN

P0-related protein (PZR), a Noonan and Leopard syndrome target, is a member of the transmembrane Immunoglobulin superfamily. Its cytoplasmic tail contains two immune-receptor tyrosine-based inhibitory motifs (ITIMs), implicated in adhesion-dependent signaling and regulating cell adhesion and motility. PZR promotes cell migration on the extracellular matrix (ECM) molecule, fibronectin, by interacting with SHP-2 (Src homology-2 domain-containing protein tyrosine phosphatase-2), a molecule essential for skeletal development and often mutated in Noonan and Leopard syndrome patients sharing overlapping musculoskeletal abnormalities and cardiac defects. To further explore the role of PZR, we assessed the expression of PZR and its ITIM-less isoform, PZRb, in human bone marrow mesenchymal stromal cells (hBM MSC), and its ability to facilitate adhesion to and spreading and migration on various ECM molecules. Furthermore, using siRNA knockdown, confocal microscopy, and immunoprecipitation assays, we assessed PZR and PZRb interactions with ß1 integrins. PZR was the predominant isoform in hBM MSC. Migrating hBM MSCs interacted most effectively with fibronectin and required the association of PZR, but not PZRb, with the integrin, VLA-5(α5ß1), leading to modulation of focal adhesion kinase phosphorylation and vinculin levels. This raises the possibility that dysregulation of PZR function may modify hBM MSC migratory behavior, potentially contributing to skeletal abnormalities.


Asunto(s)
Movimiento Celular/fisiología , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Portadoras/genética , Humanos , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA